151
|
T cell immunity to Zika virus targets immunodominant epitopes that show cross-reactivity with other Flaviviruses. Sci Rep 2018; 8:672. [PMID: 29330423 PMCID: PMC5766511 DOI: 10.1038/s41598-017-18781-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 12/18/2017] [Indexed: 11/09/2022] Open
Abstract
Zika virus (ZIKV) Infection has several outcomes from asymptomatic exposure to rash, conjunctivitis, Guillain-Barré syndrome or congenital Zika syndrome. Analysis of ZIKV immunity is confounded by the fact that several related Flaviviruses infect humans, including Dengue virus 1-4, West Nile virus and Yellow Fever virus. HLA class II restricted T cell cross-reactivity between ZIKV and other Flaviviruses infection(s) or vaccination may contribute to protection or to enhanced immunopathology. We mapped immunodominant, HLA class II restricted, CD4 epitopes from ZIKV Envelope (Env), and Non-structural (NS) NS1, NS3 and NS5 antigens in HLA class II transgenic mice. In several cases, ZIKV primed CD4 cells responded to homologous sequences from other viruses, including DENV1-4, WNV or YFV. However, cross-reactive responses could confer immune deviation - the response to the Env DENV4 p1 epitope in HLA-DR1 resulted in IL-17A immunity, often associated with exacerbated immunopathogenesis. This conservation of recognition across Flaviviruses, may encompass protective and/or pathogenic components and poses challenges to characterization of ZIKV protective immunity.
Collapse
|
152
|
Schmitt K, Charlins P, Veselinovic M, Kinner-Bibeau L, Hu S, Curlin J, Remling-Mulder L, Olson KE, Aboellail T, Akkina R. Zika viral infection and neutralizing human antibody response in a BLT humanized mouse model. Virology 2018; 515:235-242. [PMID: 29310105 DOI: 10.1016/j.virol.2017.12.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 12/21/2017] [Accepted: 12/22/2017] [Indexed: 01/27/2023]
Abstract
Many murine and non-human primate animal models have been recently developed to understand Zika viral pathogenesis. However, a major limitation with these models is the inability to directly examine the human-specific immune response. Here, we utilized a BLT humanized mouse model endowed with a transplanted human immune system. Plasma viremia could be detected within 48h after viral challenge and viremia persisted for as long as 220 days in some mice. Neutralizing human antibody was detected in infected mice and mouse sera showed reactivity with the viral envelope and capsid proteins in a radio-immunoprecipitation assay. Human monocytes/macrophages, B cells and hematopoietic stem cells in the bone marrow were found to be virus infected. These data establish that BLT mice are permissive for Zika viral infection and are capable of generating viral-specific human immune responses thus providing a human surrogate model for future testing of vaccine and antiviral therapeutic candidates.
Collapse
Affiliation(s)
- Kimberly Schmitt
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Paige Charlins
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Milena Veselinovic
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Lauren Kinner-Bibeau
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Shuang Hu
- Department of Medical Microbiology & Immunology, University of California, Davis, CA 95616, USA
| | - James Curlin
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Leila Remling-Mulder
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Ken E Olson
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Tawfik Aboellail
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Ramesh Akkina
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
153
|
Vanchiere JA, Ruiz JC, Brady AG, Kuehl TJ, Williams LE, Baze WB, Wilkerson GK, Nehete PN, McClure GB, Rogers DL, Rossi SL, Azar SR, Roundy CM, Weaver SC, Vasilakis N, Simmons JH, Abee CR. Experimental Zika Virus Infection of Neotropical Primates. Am J Trop Med Hyg 2018; 98:173-177. [PMID: 29182145 DOI: 10.4269/ajtmh.17-0322] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The establishment of a sylvatic reservoir of Zika virus (ZIKV) in the Americas is dependent on the susceptibility of primates of sufficient population density, the duration and magnitude of viremia, and their exposure to the human mosquito-borne transmission cycle. To assess the susceptibility of squirrel (Saimiri sp.) and owl monkeys (Aotus sp.) to infection, we inoculated four animals of each species with ZIKV from the current epidemic. Viremia in the absence of detectible disease was observed in both species and seroconversion occurred by day 28. ZIKV was detected in the spleen of three owl monkeys: one at 7 days postinoculation (dpi) and two at 14 dpi. This study confirms the susceptibility to ZIKV infection of two Neotropical primate species that live in close proximity to humans in South America, suggesting that they could support a widespread sylvatic ZIKV cycle there. Collectively, establishment of a ZIKV sylvatic transmission cycle in South America would imperil eradication efforts and could provide a mechanism for continued exposure of humans to ZIKV infection and disease.
Collapse
Affiliation(s)
- John A Vanchiere
- Department of Veterinary Sciences, The University of Texas MD Anderson Cancer Center, Bastrop, Texas.,Department of Pediatrics, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Julio C Ruiz
- Department of Veterinary Sciences, The University of Texas MD Anderson Cancer Center, Bastrop, Texas
| | - Alan G Brady
- Department of Veterinary Sciences, The University of Texas MD Anderson Cancer Center, Bastrop, Texas
| | - Thomas J Kuehl
- Department of Obstetrics and Gynecology, Baylor Scott and White Clinic, Temple, Texas
| | - Lawrence E Williams
- Department of Veterinary Sciences, The University of Texas MD Anderson Cancer Center, Bastrop, Texas
| | - Wallace B Baze
- Department of Veterinary Sciences, The University of Texas MD Anderson Cancer Center, Bastrop, Texas
| | - Gregory K Wilkerson
- Department of Veterinary Sciences, The University of Texas MD Anderson Cancer Center, Bastrop, Texas
| | - Pramod N Nehete
- Department of Veterinary Sciences, The University of Texas MD Anderson Cancer Center, Bastrop, Texas
| | - Gloria B McClure
- Department of Pediatrics, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Donna L Rogers
- Department of Pediatrics, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Shannan L Rossi
- Department of Pathology and Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas
| | - Sasha R Azar
- Department of Pathology and Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas
| | - Christopher M Roundy
- Department of Pathology and Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas
| | - Scott C Weaver
- Department of Microbiology and Immunology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas
| | - Nikos Vasilakis
- Department of Microbiology and Immunology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas.,Department of Pathology and Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas
| | - Joe H Simmons
- Department of Veterinary Sciences, The University of Texas MD Anderson Cancer Center, Bastrop, Texas
| | - Christian R Abee
- Department of Veterinary Sciences, The University of Texas MD Anderson Cancer Center, Bastrop, Texas
| |
Collapse
|
154
|
Gallichotte EN, Dinnon KH, Lim XN, Ng TS, Lim EXY, Menachery VD, Lok SM, Baric RS. CD-loop Extension in Zika Virus Envelope Protein Key for Stability and Pathogenesis. J Infect Dis 2017; 216:1196-1204. [PMID: 28968838 DOI: 10.1093/infdis/jix473] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 09/06/2017] [Indexed: 11/14/2022] Open
Abstract
With severe disease manifestations including microcephaly, congenital malformation, and Guillain-Barré syndrome, Zika virus (ZIKV) remains a persistent global public health threat. Despite antigenic similarities with dengue viruses, structural studies have suggested the extended CD-loop and hydrogen-bonding interaction network within the ZIKV envelope protein contribute to stability differences between the viral families. This enhanced stability may lead to the augmented infection, disease manifestation, and persistence in body fluids seen following ZIKV infection. To examine the role of these motifs in infection, we generated a series of ZIKV recombinant viruses that disrupted the hydrogen-bonding network (350A, 351A, and 350A/351A) or the CD-loop extension (Δ346). Our results demonstrate a key role for the ZIKV extended CD-loop in cell-type-dependent replication, virion stability, and in vivo pathogenesis. Importantly, the Δ346 mutant maintains similar antigenicity to wild-type virus, opening the possibility for its use as a live-attenuated vaccine platform for ZIKV and other clinically relevant flaviviruses.
Collapse
Affiliation(s)
- Emily N Gallichotte
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill
| | - Kenneth H Dinnon
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill
| | - Xin-Ni Lim
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Graduate Medical School.,Centre for BioImaging Science, National University of Singapore
| | - Thiam-Seng Ng
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Graduate Medical School.,Centre for BioImaging Science, National University of Singapore
| | - Elisa X Y Lim
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Graduate Medical School.,Centre for BioImaging Science, National University of Singapore
| | - Vineet D Menachery
- Department of Epidemiology, University of North Carolina at Chapel Hill.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston
| | - Shee-Mei Lok
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Graduate Medical School.,Centre for BioImaging Science, National University of Singapore
| | - Ralph S Baric
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill.,Department of Epidemiology, University of North Carolina at Chapel Hill
| |
Collapse
|
155
|
Ricciardi MJ, Magnani DM, Grifoni A, Kwon YC, Gutman MJ, Grubaugh ND, Gangavarapu K, Sharkey M, Silveira CGT, Bailey VK, Pedreño-Lopez N, Gonzalez-Nieto L, Maxwell HS, Domingues A, Martins MA, Pham J, Weiskopf D, Altman J, Kallas EG, Andersen KG, Stevenson M, Lichtenberger P, Choe H, Whitehead SS, Sette A, Watkins DI. Ontogeny of the B- and T-cell response in a primary Zika virus infection of a dengue-naïve individual during the 2016 outbreak in Miami, FL. PLoS Negl Trop Dis 2017; 11:e0006000. [PMID: 29267278 PMCID: PMC5755934 DOI: 10.1371/journal.pntd.0006000] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 01/05/2018] [Accepted: 09/28/2017] [Indexed: 01/05/2023] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus of significant public health concern. In the summer of 2016, ZIKV was first detected in the contiguous United States. Here we present one of the first cases of a locally acquired ZIKV infection in a dengue-naïve individual. We collected blood from a female with a maculopapular rash at day (D) 5 and D7 post onset of symptoms (POS) and we continued weekly blood draws out to D148 POS. To establish the ontogeny of the immune response against ZIKV, lymphocytes and plasma were analyzed in a longitudinal fashion. The plasmablast response peaked at D7 POS (19.6% of CD19+ B-cells) and was undetectable by D15 POS. ZIKV-specific IgM was present at D5 POS, peaked between D15 and D21 POS, and subsequently decreased. The ZIKV-specific IgG response, however, was not detected until D15 POS and continued to increase after that. Interestingly, even though the patient had never been infected with dengue virus (DENV), cross-reactive IgM and IgG binding against each of the four DENV serotypes could be detected. The highest plasma neutralization activity against ZIKV peaked between D15 and D21 POS, and even though DENV binding antibodies were present in the plasma of the patient, there was neither neutralization nor antibody dependent enhancement (ADE) of DENV. Interestingly, ADE against ZIKV arose at D48 POS and continued until the end of the study. CD4+ and CD8+ T-cells recognized ZIKV-NS2A and ZIKV-E, respectively. The tetramer positive CD8+ T-cell response peaked at D21 POS with elevated levels persisting for months. In summary, this is the first study to establish the timing of the ontogeny of the immune response against ZIKV. Zika virus (ZIKV) is an emerging viral disease that has the potential to negatively impact future generations by causing birth defects in infected pregnant mothers. While there have been many studies performed in animal models of ZIKV infection, there have only been a limited number of reports studying the immune responses in humans. Ricciardi et. al. analyzed the immune response of a primary ZIKV infection in a dengue virus (DENV) naïve individual during the 2016 outbreak in Miami, Florida. B- and T-cell responses were assessed over multiple time points. Cross-reactive antibodies against DENV, a virus that the patient was never infected with, were generated during the ZIKV infection, but these antibodies failed to neutralize any of the DENV serotypes. Furthermore, while these DENV-cross-reactive antibodies might be expected to cause antibody dependent enhancement (ADE) of DENV infection, they did not. Interestingly, ADE of ZIKV infection was seen at approximately 1 ½ months after infection. Together, these results establish the timing of the ontogeny of the immune response against a primary ZIKV infection in a DENV-naïve individual.
Collapse
Affiliation(s)
- Michael J. Ricciardi
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, United States of America
- * E-mail:
| | - Diogo M. Magnani
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Alba Grifoni
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States of America
| | - Young-Chan Kwon
- Department of Immunology and Microbial Science, The Scripps Research Institute, Jupiter, FL, United States of America
| | - Martin J. Gutman
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Nathan D. Grubaugh
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, United States of America
| | - Karthik Gangavarapu
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, United States of America
| | - Mark Sharkey
- Division of Infectious Disease, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Cassia G. T. Silveira
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | - Varian K. Bailey
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Núria Pedreño-Lopez
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Lucas Gonzalez-Nieto
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Helen S. Maxwell
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Aline Domingues
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Mauricio A. Martins
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - John Pham
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States of America
| | - Daniela Weiskopf
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States of America
| | - John Altman
- Department of Microbiology and Immunology and Emory Vaccine Research Center, Emory University, Atlanta, GA, United States of America
| | - Esper G. Kallas
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | - Kristian G. Andersen
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, United States of America
| | - Mario Stevenson
- Division of Infectious Disease, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Paola Lichtenberger
- Division of Infectious Disease, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Hyeryun Choe
- Department of Immunology and Microbial Science, The Scripps Research Institute, Jupiter, FL, United States of America
| | - Stephen S. Whitehead
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States of America
| | - David I. Watkins
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, United States of America
| |
Collapse
|
156
|
Dudley DM, Newman CM, Lalli J, Stewart LM, Koenig MR, Weiler AM, Semler MR, Barry GL, Zarbock KR, Mohns MS, Breitbach ME, Schultz-Darken N, Peterson E, Newton W, Mohr EL, Capuano Iii S, Osorio JE, O'Connor SL, O'Connor DH, Friedrich TC, Aliota MT. Infection via mosquito bite alters Zika virus tissue tropism and replication kinetics in rhesus macaques. Nat Commun 2017; 8:2096. [PMID: 29235456 PMCID: PMC5727388 DOI: 10.1038/s41467-017-02222-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/13/2017] [Indexed: 11/24/2022] Open
Abstract
Mouse and nonhuman primate models now serve as useful platforms to study Zika virus (ZIKV) pathogenesis, candidate therapies, and vaccines, but they rely on needle inoculation of virus: the effects of mosquito-borne infection on disease outcome have not been explored in these models. Here we show that infection via mosquito bite delays ZIKV replication to peak viral loads in rhesus macaques. Importantly, in mosquito-infected animals ZIKV tissue distribution was limited to hemolymphatic tissues, female reproductive tract tissues, kidney, and liver, potentially emulating key features of human ZIKV infections, most of which are characterized by mild or asymptomatic disease. Furthermore, deep sequencing analysis reveals that ZIKV populations in mosquito-infected monkeys show greater sequence heterogeneity and lower overall diversity than in needle-inoculated animals. This newly developed system will be valuable for studying ZIKV disease because it more closely mimics human infection by mosquito bite than needle-based inoculations. Vector saliva can affect infectivity and pathogenesis of vector-borne viruses, but this hasn’t been studied for Zika virus infection. Here, Dudley et al. show that mosquito-mediated Zika infection of macaques results in altered replication kinetics and greater sequence heterogeneity.
Collapse
Affiliation(s)
- Dawn M Dudley
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 3170 UW Medical Foundation Centennial Building, 1685 Highland Ave., Madison, WI, 53705, USA
| | - Christina M Newman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 3170 UW Medical Foundation Centennial Building, 1685 Highland Ave., Madison, WI, 53705, USA
| | - Joseph Lalli
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, 1656 Linden Dr., Madison, WI, 53706, USA
| | - Laurel M Stewart
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 3170 UW Medical Foundation Centennial Building, 1685 Highland Ave., Madison, WI, 53705, USA
| | - Michelle R Koenig
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 3170 UW Medical Foundation Centennial Building, 1685 Highland Ave., Madison, WI, 53705, USA
| | - Andrea M Weiler
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1220 Capitol Ct., Madison, WI, 53715, USA
| | - Matthew R Semler
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 3170 UW Medical Foundation Centennial Building, 1685 Highland Ave., Madison, WI, 53705, USA
| | - Gabrielle L Barry
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1220 Capitol Ct., Madison, WI, 53715, USA
| | - Katie R Zarbock
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 3170 UW Medical Foundation Centennial Building, 1685 Highland Ave., Madison, WI, 53705, USA
| | - Mariel S Mohns
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 3170 UW Medical Foundation Centennial Building, 1685 Highland Ave., Madison, WI, 53705, USA
| | - Meghan E Breitbach
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 3170 UW Medical Foundation Centennial Building, 1685 Highland Ave., Madison, WI, 53705, USA
| | - Nancy Schultz-Darken
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1220 Capitol Ct., Madison, WI, 53715, USA
| | - Eric Peterson
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1220 Capitol Ct., Madison, WI, 53715, USA
| | - Wendy Newton
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1220 Capitol Ct., Madison, WI, 53715, USA
| | - Emma L Mohr
- Department of Pediatrics, University of Wisconsin-Madison, University of Wisconsin Clinical Science Center, 600 Highland Ave, Madison, WI, 53792, USA
| | - Saverio Capuano Iii
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1220 Capitol Ct., Madison, WI, 53715, USA
| | - Jorge E Osorio
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, 1656 Linden Dr., Madison, WI, 53706, USA
| | - Shelby L O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 3170 UW Medical Foundation Centennial Building, 1685 Highland Ave., Madison, WI, 53705, USA.,Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1220 Capitol Ct., Madison, WI, 53715, USA
| | - David H O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 3170 UW Medical Foundation Centennial Building, 1685 Highland Ave., Madison, WI, 53705, USA.,Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1220 Capitol Ct., Madison, WI, 53715, USA
| | - Thomas C Friedrich
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, 1656 Linden Dr., Madison, WI, 53706, USA.,Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1220 Capitol Ct., Madison, WI, 53715, USA
| | - Matthew T Aliota
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, 1656 Linden Dr., Madison, WI, 53706, USA.
| |
Collapse
|
157
|
Abbink P, Larocca RA, Visitsunthorn K, Boyd M, De La Barrera RA, Gromowski GD, Kirilova M, Peterson R, Li Z, Nanayakkara O, Nityanandam R, Mercado NB, Borducchi EN, Chandrashekar A, Jetton D, Mojta S, Gandhi P, LeSuer J, Khatiwada S, Lewis MG, Modjarrad K, Jarman RG, Eckels KH, Thomas SJ, Michael NL, Barouch DH. Durability and correlates of vaccine protection against Zika virus in rhesus monkeys. Sci Transl Med 2017; 9:eaao4163. [PMID: 29237759 PMCID: PMC5747972 DOI: 10.1126/scitranslmed.aao4163] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/07/2017] [Accepted: 11/22/2017] [Indexed: 12/22/2022]
Abstract
An effective Zika virus (ZIKV) vaccine will require long-term durable protection. Several ZIKV vaccine candidates have demonstrated protective efficacy in nonhuman primates, but these studies have typically involved ZIKV challenge shortly after vaccination at peak immunity. We show that a single immunization with an adenovirus vector-based vaccine, as well as two immunizations with a purified inactivated virus vaccine, afforded robust protection against ZIKV challenge in rhesus monkeys at 1 year after vaccination. In contrast, two immunizations with an optimized DNA vaccine, which provided complete protection at peak immunity, resulted in reduced protective efficacy at 1 year that was associated with declining neutralizing antibody titers to subprotective levels. These data define a microneutralization log titer of 2.0 to 2.1 as the threshold required for durable protection against ZIKV challenge in this model. Moreover, our findings demonstrate that protection against ZIKV challenge in rhesus monkeys is possible for at least 1 year with a single-shot vaccine.
Collapse
Affiliation(s)
- Peter Abbink
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Rafael A Larocca
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Kittipos Visitsunthorn
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Michael Boyd
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | | - Marinela Kirilova
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Rebecca Peterson
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Zhenfeng Li
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ovini Nanayakkara
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ramya Nityanandam
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Noe B Mercado
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Erica N Borducchi
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Abishek Chandrashekar
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - David Jetton
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Shanell Mojta
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Priya Gandhi
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jake LeSuer
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Shreeya Khatiwada
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | - Kayvon Modjarrad
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Richard G Jarman
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Kenneth H Eckels
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Stephen J Thomas
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Nelson L Michael
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA 02139, USA
| |
Collapse
|
158
|
Blackman MA, Kim IJ, Lin JS, Thomas SJ. Challenges of Vaccine Development for Zika Virus. Viral Immunol 2017; 31:117-123. [PMID: 29227202 DOI: 10.1089/vim.2017.0145] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The emergence of outbreaks of Zika virus (ZIKV) in Brazil in 2015 was associated with devastating effects on fetal development and prompted a world health emergency and multiple efforts to generate an effective vaccine against infection. There are now more than 40 vaccine candidates in preclinical development and six in clinical trials. Despite similarities with other flaviviruses to which successful vaccines have been developed, such as yellow fever virus and Japanese Encephalitis virus, there are unique challenges to the development and clinical trials of a vaccine for ZIKV.
Collapse
Affiliation(s)
| | | | | | - Stephen J Thomas
- 2 Infectious Disease Division, Upstate Medical University, State University of New York , Syracuse, New York
| |
Collapse
|
159
|
Chiu CY, Sánchez-San Martín C, Bouquet J, Li T, Yagi S, Tamhankar M, Hodara VL, Parodi LM, Somasekar S, Yu G, Giavedoni LD, Tardif S, Patterson J. Experimental Zika Virus Inoculation in a New World Monkey Model Reproduces Key Features of the Human Infection. Sci Rep 2017; 7:17126. [PMID: 29215081 PMCID: PMC5719425 DOI: 10.1038/s41598-017-17067-w] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/16/2017] [Indexed: 11/13/2022] Open
Abstract
A monkey model of Zika virus (ZIKV) infection is urgently needed to better understand transmission and pathogenesis, given its proven association with fetal brain defects in pregnant women and acute neurological illness. Here we experimentally infected 4 male marmosets with ZIKV (prototype 1947 African strain) and monitored them clinically with sampling of various body fluids and tissues for nearly 3 months. We show that the course of acute infection with ZIKV in these New World monkeys resembles the human illness in many respects, including (1) lack of apparent clinical symptoms in most cases, (2) persistence of the virus in body fluids such as semen and saliva for longer periods of time than in serum, and (3) generation of neutralizing antibodies as well as an antiviral immunological host response. Importantly, ZIKV-infected saliva samples (in addition to serum) were found to be infectious, suggesting potential capacity for viral transmission by the oral route. Re-challenge of a previously infected marmoset with a contemporary outbreak strain SPH2015 from Brazil resulted in continued protection against infection, no viral shedding, and boosting of the immune response. Given the key similarities to human infection, a marmoset model of ZIKV infection may be useful for testing of new drugs and vaccines.
Collapse
Affiliation(s)
- Charles Y Chiu
- Department of Laboratory Medicine, University of California, San Francisco, CA, 94107, USA. .,UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, 91407, USA. .,Department of Medicine, Division of Infectious Diseases, University of California, San Francisco, CA, 94107, USA.
| | - Claudia Sánchez-San Martín
- Department of Laboratory Medicine, University of California, San Francisco, CA, 94107, USA.,UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, 91407, USA
| | - Jerome Bouquet
- Department of Laboratory Medicine, University of California, San Francisco, CA, 94107, USA.,UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, 91407, USA
| | - Tony Li
- Department of Laboratory Medicine, University of California, San Francisco, CA, 94107, USA.,UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, 91407, USA
| | - Shigeo Yagi
- California Department of Public Health, Richmond, CA, USA
| | | | - Vida L Hodara
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Laura M Parodi
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Sneha Somasekar
- Department of Laboratory Medicine, University of California, San Francisco, CA, 94107, USA.,UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, 91407, USA
| | - Guixia Yu
- Department of Laboratory Medicine, University of California, San Francisco, CA, 94107, USA.,UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, 91407, USA
| | | | - Suzette Tardif
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Jean Patterson
- Texas Biomedical Research Institute, San Antonio, TX, USA
| |
Collapse
|
160
|
Thompson SJ, Pearce JM, Ramey AM. Vectors, Hosts, and Control Measures for Zika Virus in the Americas. ECOHEALTH 2017; 14:821-839. [PMID: 29150828 DOI: 10.1007/s10393-017-1277-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 08/11/2017] [Accepted: 08/14/2017] [Indexed: 06/07/2023]
Abstract
We examine Zika virus (ZIKV) from an ecological perspective and with a focus on the Americas. We assess (1) the role of wildlife in ZIKV disease ecology, (2) how mosquito behavior and biology influence disease dynamics, and (3) how nontarget species and ecosystems may be impacted by vector control programs. Our review suggests that free-ranging, non-human primates may be involved in ZIKV transmission in the Old World; however, other wildlife species likely play a limited role in maintaining or transmitting ZIKV. In the Americas, a zoonotic cycle has not yet been definitively established. Understanding behaviors and habitat tolerances of Aedes aegypti and Aedes albopictus, two ZIKV competent vectors in the Americas, will allow more accurate modeling of disease spread and facilitate targeted and effective control efforts. Vector control efforts may have direct and indirect impacts to wildlife, particularly invertebrate feeding species; however, strategies could be implemented to limit detrimental ecological effects.
Collapse
Affiliation(s)
- Sarah J Thompson
- U.S. Geological Survey, Alaska Science Center, 4210 University Drive, Anchorage, AK, USA.
| | - John M Pearce
- U.S. Geological Survey, Alaska Science Center, 4210 University Drive, Anchorage, AK, USA
| | - Andrew M Ramey
- U.S. Geological Survey, Alaska Science Center, 4210 University Drive, Anchorage, AK, USA
| |
Collapse
|
161
|
Javed F, Manzoor KN, Ali M, Haq IU, Khan AA, Zaib A, Manzoor S. Zika virus: what we need to know? J Basic Microbiol 2017; 58:3-16. [DOI: 10.1002/jobm.201700398] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 09/19/2017] [Accepted: 09/03/2017] [Indexed: 01/22/2023]
Affiliation(s)
- Farakh Javed
- Department of Microbiology; University of Haripur; Haripur Pakistan
| | | | - Mubashar Ali
- Department of Microbiology; University of Haripur; Haripur Pakistan
| | - Irshad U. Haq
- Department of Microbiology; University of Haripur; Haripur Pakistan
| | - Abid A. Khan
- Department of Biosciences; COMSATS Institute of Information Technology; Islamabad Pakistan
| | - Assad Zaib
- Department of Medical Lab Technology; University of Haripur; Haripur Pakistan
| | - Sobia Manzoor
- Atta-ur-Rehman School of Applied Bio-Sciences; National University of Science and Technology; Islamabad Pakistan
| |
Collapse
|
162
|
Fernandez E, Dejnirattisai W, Cao B, Scheaffer SM, Supasa P, Wongwiwat W, Esakky P, Drury A, Mongkolsapaya J, Moley KH, Mysorekar IU, Screaton GR, Diamond MS. Human antibodies to the dengue virus E-dimer epitope have therapeutic activity against Zika virus infection. Nat Immunol 2017; 18:1261-1269. [PMID: 28945244 PMCID: PMC5679314 DOI: 10.1038/ni.3849] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 09/01/2017] [Indexed: 12/12/2022]
Abstract
The Zika virus (ZIKV) epidemic has resulted in congenital abnormalities in fetuses and neonates. Although some cross-reactive dengue virus (DENV)-specific antibodies can enhance ZIKV infection in mice, those recognizing the DENV E-dimer epitope (EDE) can neutralize ZIKV infection in cell culture. We evaluated the therapeutic activity of human monoclonal antibodies to DENV EDE for their ability to control ZIKV infection in the brains, testes, placentas, and fetuses of mice. A single dose of the EDE1-B10 antibody given 3 d after ZIKV infection protected against lethality, reduced ZIKV levels in brains and testes, and preserved sperm counts. In pregnant mice, wild-type or engineered LALA variants of EDE1-B10, which cannot engage Fcg receptors, diminished ZIKV burden in maternal and fetal tissues, and protected against fetal demise. Because neutralizing antibodies to EDE have therapeutic potential against ZIKV, in addition to their established inhibitory effects against DENV, it may be possible to develop therapies that control disease caused by both viruses.
Collapse
Affiliation(s)
- Estefania Fernandez
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Wanwisa Dejnirattisai
- Division of Immunology and Inflammation, Department of Medicine, Hammersmith Campus, Imperial College London, UK
| | - Bin Cao
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St Louis, MO, USA
| | - Suzanne M. Scheaffer
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St Louis, MO, USA
| | - Piyada Supasa
- Division of Immunology and Inflammation, Department of Medicine, Hammersmith Campus, Imperial College London, UK
| | - Wiyada Wongwiwat
- Division of Immunology and Inflammation, Department of Medicine, Hammersmith Campus, Imperial College London, UK
| | - Prabagaran Esakky
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St Louis, MO, USA
| | - Andrea Drury
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St Louis, MO, USA
| | - Juthathip Mongkolsapaya
- Division of Immunology and Inflammation, Department of Medicine, Hammersmith Campus, Imperial College London, UK
- Dengue Hemorrhagic Fever Research Unit, Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol University, Bangkok, Thailand
| | - Kelle H. Moley
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St Louis, MO, USA
| | - Indira U. Mysorekar
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO, USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St Louis, MO, USA
| | - Gavin R. Screaton
- Division of Immunology and Inflammation, Department of Medicine, Hammersmith Campus, Imperial College London, UK
| | - Michael S. Diamond
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
163
|
Abstract
Here, Wen et al. review the current knowledge and progress in understanding the impact of Zika virus exposure on mammalian brain development and discuss potential underlying mechanisms. The re-emergence of Zika virus (ZIKV), a mosquito-borne and sexually transmitted flavivirus circulating in >70 countries and territories, poses a significant global threat to public health due to its ability to cause severe developmental defects in the human brain, such as microcephaly. Since the World Health Organization declared the ZIKV outbreak a Public Health Emergency of International Concern, remarkable progress has been made to gain insight into cellular targets, pathogenesis, and underlying biological mechanisms of ZIKV infection. Here we review the current knowledge and progress in understanding the impact of ZIKV exposure on the mammalian brain development and discuss potential underlying mechanisms.
Collapse
Affiliation(s)
- Zhexing Wen
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA.,Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia 30322, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Hongjun Song
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.,The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.,Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Guo-Li Ming
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.,The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.,Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
164
|
Hirsch AJ. Modeling Zika virus infection in nonhuman primates. Future Virol 2017. [DOI: 10.2217/fvl-2017-0067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Alec J Hirsch
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| |
Collapse
|
165
|
Villamil-Gómez WE, Guijarro E, Castellanos J, Rodríguez-Morales AJ. Congenital Zika syndrome with prolonged detection of Zika virus RNA. J Clin Virol 2017; 95:52-54. [PMID: 28866135 DOI: 10.1016/j.jcv.2017.08.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 06/13/2017] [Accepted: 08/21/2017] [Indexed: 01/23/2023]
Affiliation(s)
- Wilmer E Villamil-Gómez
- Infectious Diseases and Infection Control Research Group, Hospital Universitario de Sincelejo, Sincelejo, Sucre, Colombia; Programa del Doctorado de Medicina Tropical, SUE Caribe, Universidad del Atlántico, Barranquilla, Colombia
| | | | | | - Alfonso J Rodríguez-Morales
- Public Health and Infection Group of Research, Faculty of Health Sciences, Universidad Tecnológica de Pereira, Pereira, Risaralda, Colombia.
| |
Collapse
|
166
|
Ferreira AC, Zaverucha-do-Valle C, Reis PA, Barbosa-Lima G, Vieira YR, Mattos M, Silva PDP, Sacramento C, de Castro Faria Neto HC, Campanati L, Tanuri A, Brüning K, Bozza FA, Bozza PT, Souza TML. Sofosbuvir protects Zika virus-infected mice from mortality, preventing short- and long-term sequelae. Sci Rep 2017; 7:9409. [PMID: 28842610 PMCID: PMC5573375 DOI: 10.1038/s41598-017-09797-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 07/28/2017] [Indexed: 01/04/2023] Open
Abstract
Zika virus (ZIKV) causes significant public health concerns because of its association with congenital malformations, neurological disorders in adults, and, more recently, death. Considering the necessity to mitigate ZIKV-associated diseases, antiviral interventions are an urgent necessity. Sofosbuvir, a drug in clinical use against hepatitis C virus (HCV), is among the FDA-approved substances endowed with anti-ZIKV activity. In this work, we further investigated the in vivo activity of sofosbuvir against ZIKV. Neonatal Swiss mice were infected with ZIKV (2 × 107 PFU) and treated with sofosbuvir at 20 mg/kg/day, a concentration compatible with pre-clinical development of this drug. We found that sofosbuvir reduced acute levels of ZIKV from 60 to 90% in different anatomical compartments, such as the blood plasma, spleen, kidney, and brain. Early treatment with sofosbuvir doubled the percentage and time of survival of ZIKV-infected animals. Sofosbuvir also prevented the acute neuromotor impairment triggered by ZIKV. In the long-term behavioural analysis of ZIKV-associated sequelae, sofosbuvir prevented loss of hippocampal- and amygdala-dependent memory. Our results indicate that sofosbuvir inhibits ZIKV replication in vivo, which is consistent with the prospective necessity of antiviral drugs to treat ZIKV-infected individuals.
Collapse
Affiliation(s)
- André C Ferreira
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Camila Zaverucha-do-Valle
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- Instituto Nacional de Infectologia (INI), Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Patrícia A Reis
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | | | | | - Mayara Mattos
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Priscila de Paiva Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Carolina Sacramento
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Hugo C de Castro Faria Neto
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Loraine Campanati
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Amilcar Tanuri
- Instituto de Biologia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Karin Brüning
- BMK Consortium: Blanver Farmoquímica Ltda; Microbiológica Química e Farmacêutica Ltda, Karin Bruning & Cia. Ltda, Taboão da Serra, SP, Brazil
| | - Fernando A Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- Instituto Nacional de Infectologia (INI), Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Patrícia T Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Thiago Moreno L Souza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil.
- Instituto Nacional de Infectologia (INI), Fiocruz, Rio de Janeiro, RJ, Brazil.
- National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Fiocruz, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
167
|
Abstract
The 2015 Brazilian Zika virus outbreak sparked a rapid response to control the spread of the virus. What was first understood to be a mild self-resolving infection is now linked to significant neurological defects in both neonates and adults. The WHO declared the 2016 Zika epidemic a public health emergency and issued an unprecedented recommendation to women in affected regions to delay pregnancy until the risks surrounding Zika virus could be understood, or the epidemic contained. Since that time, considerable effort has been dedicated to understanding Zika transmission and pathogenesis to aid the development of drugs and vaccines. Several models have emerged to study numerous facets of Zika biology; this review details the various model systems.
Collapse
|
168
|
Munjal A, Khandia R, Dhama K, Sachan S, Karthik K, Tiwari R, Malik YS, Kumar D, Singh RK, Iqbal HMN, Joshi SK. Advances in Developing Therapies to Combat Zika Virus: Current Knowledge and Future Perspectives. Front Microbiol 2017; 8:1469. [PMID: 28824594 PMCID: PMC5541032 DOI: 10.3389/fmicb.2017.01469] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 07/20/2017] [Indexed: 02/05/2023] Open
Abstract
Zika virus (ZIKV) remained largely quiescent for nearly six decades after its first appearance in 1947. ZIKV reappeared after 2007, resulting in a declaration of an international "public health emergency" in 2016 by the World Health Organization (WHO). Until this time, ZIKV was considered to induce only mild illness, but it has now been established as the cause of severe clinical manifestations, including fetal anomalies, neurological problems, and autoimmune disorders. Infection during pregnancy can cause congenital brain abnormalities, including microcephaly and neurological degeneration, and in other cases, Guillain-Barré syndrome, making infections with ZIKV a substantial public health concern. Genomic and molecular investigations are underway to investigate ZIKV pathology and its recent enhanced pathogenicity, as well as to design safe and potent vaccines, drugs, and therapeutics. This review describes progress in the design and development of various anti-ZIKV therapeutics, including drugs targeting virus entry into cells and the helicase protein, nucleosides, inhibitors of NS3 protein, small molecules, methyltransferase inhibitors, interferons, repurposed drugs, drugs designed with the aid of computers, neutralizing antibodies, convalescent serum, antibodies that limit antibody-dependent enhancement, and herbal medicines. Additionally, covalent inhibitors of viral protein expression and anti-Toll-like receptor molecules are discussed. To counter ZIKV-associated disease, we need to make rapid progress in developing novel therapies that work effectually to inhibit ZIKV.
Collapse
Affiliation(s)
- Ashok Munjal
- Department of Biochemistry and Genetics, Barkatullah UniversityBhopal, India
| | - Rekha Khandia
- Department of Biochemistry and Genetics, Barkatullah UniversityBhopal, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research InstituteBareilly, India
| | - Swati Sachan
- Immunology Section, ICAR-Indian Veterinary Research InstituteBareilly, India
| | - Kumaragurubaran Karthik
- Central University Laboratory, Tamil Nadu Veterinary and Animal Sciences UniversityChennai, India
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, UP Pandit Deen Dayal Upadhayay Pashu Chikitsa Vigyan Vishwavidyalay Evum Go-Anusandhan SansthanMathura, India
| | - Yashpal S. Malik
- Division of Biological Standardization, ICAR-Indian Veterinary Research InstituteBareilly, India
| | - Deepak Kumar
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research InstituteBareilly, India
| | - Raj K. Singh
- ICAR-Indian Veterinary Research InstituteBareilly, India
| | - Hafiz M. N. Iqbal
- School of Engineering and Science, Tecnologico de Monterrey, Campus MonterreyMonterrey, Mexico
| | - Sunil K. Joshi
- Cellular Immunology Lab, Frank Reidy Research Center of Bioelectrics, Old Dominion University, NorfolkVA, United States
| |
Collapse
|
169
|
Newman C, Friedrich TC, O'Connor DH. Macaque monkeys in Zika virus research: 1947-present. Curr Opin Virol 2017; 25:34-40. [PMID: 28750247 PMCID: PMC5610623 DOI: 10.1016/j.coviro.2017.06.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 06/19/2017] [Accepted: 06/22/2017] [Indexed: 02/06/2023]
Abstract
Zika virus was first isolated in 1947 from an exotic rhesus macaque. Nearly 70 years later, the emergence of Zika virus in the Americas and its newly described association with birth defects has motivated the development of captive macaque monkey models of human Zika virus infection. This review describes similarities between macaque and human Zika virus pathogenesis and discusses specific advantages and disadvantages of using macaques instead of other laboratory animal models. In particular, macaques provide an outstanding model for understanding in utero Zika virus infections that are essential for evaluating preclinical interventions for use in pregnancy.
Collapse
Affiliation(s)
- Christina Newman
- University of Wisconsin-Madison Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, 3170 Medical Foundation Centennial Building (MFCB), 1685 Highland Avenue, Madison, WI 53705, United States
| | - Thomas C Friedrich
- University of Wisconsin-Madison Department of Pathobiological Sciences, School of Veterinary Medicine, 2015 Linden Drive, Madison, WI 53706, United States; Wisconsin National Primate Research Center (WNPRC), 1220 Capitol Court, Madison, WI 53715, United States
| | - David H O'Connor
- University of Wisconsin-Madison Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, 3170 Medical Foundation Centennial Building (MFCB), 1685 Highland Avenue, Madison, WI 53705, United States; Wisconsin National Primate Research Center (WNPRC), 1220 Capitol Court, Madison, WI 53715, United States.
| |
Collapse
|
170
|
Aliota MT, Bassit L, Bradrick SS, Cox B, Garcia-Blanco MA, Gavegnano C, Friedrich TC, Golos TG, Griffin DE, Haddow AD, Kallas EG, Kitron U, Lecuit M, Magnani DM, Marrs C, Mercer N, McSweegan E, Ng LFP, O'Connor DH, Osorio JE, Ribeiro GS, Ricciardi M, Rossi SL, Saade G, Schinazi RF, Schott-Lerner GO, Shan C, Shi PY, Watkins DI, Vasilakis N, Weaver SC. Zika in the Americas, year 2: What have we learned? What gaps remain? A report from the Global Virus Network. Antiviral Res 2017; 144:223-246. [PMID: 28595824 PMCID: PMC5920658 DOI: 10.1016/j.antiviral.2017.06.001] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 06/01/2017] [Indexed: 12/25/2022]
Abstract
In response to the outbreak of Zika virus (ZIKV) infection in the Western Hemisphere and the recognition of a causal association with fetal malformations, the Global Virus Network (GVN) assembled an international taskforce of virologists to promote basic research, recommend public health measures and encourage the rapid development of vaccines, antiviral therapies and new diagnostic tests. In this article, taskforce members and other experts review what has been learned about ZIKV-induced disease in humans, its modes of transmission and the cause and nature of associated congenital manifestations. After describing the make-up of the taskforce, we summarize the emergence of ZIKV in the Americas, Africa and Asia, its spread by mosquitoes, and current control measures. We then review the spectrum of primary ZIKV-induced disease in adults and children, sites of persistent infection and sexual transmission, then examine what has been learned about maternal-fetal transmission and the congenital Zika syndrome, including knowledge obtained from studies in laboratory animals. Subsequent sections focus on vaccine development, antiviral therapeutics and new diagnostic tests. After reviewing current understanding of the mechanisms of emergence of Zika virus, we consider the likely future of the pandemic.
Collapse
Affiliation(s)
- Matthew T Aliota
- Department of Pathobiological Sciences, University of Wisconsin-Madison, USA
| | - Leda Bassit
- Center for AIDS Research, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Shelton S Bradrick
- Department of Biochemistry and Molecular Biology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Bryan Cox
- Center for AIDS Research, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Mariano A Garcia-Blanco
- Department of Biochemistry and Molecular Biology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Christina Gavegnano
- Center for AIDS Research, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Thomas C Friedrich
- Department of Pathobiological Sciences, University of Wisconsin-Madison, USA; Wisconsin National Primate Research Center, University of Wisconsin-Madison, USA
| | - Thaddeus G Golos
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, USA; Department of Comparative Biosciences, University of Wisconsin-Madison, USA; Department of Obstetrics and Gynecology, University of Wisconsin-Madison, USA
| | - Diane E Griffin
- Global Virus Network, 725 West Lombard St., Baltimore, MD, USA; W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Andrew D Haddow
- Global Virus Network, 725 West Lombard St., Baltimore, MD, USA; Virology Division, United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD, 21702, USA
| | - Esper G Kallas
- Global Virus Network, 725 West Lombard St., Baltimore, MD, USA; Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, Brazil
| | - Uriel Kitron
- Department of Environmental Sciences, Emory University, Atlanta, GA, USA
| | - Marc Lecuit
- Global Virus Network, 725 West Lombard St., Baltimore, MD, USA; Institut Pasteur, Biology of Infection Unit and INSERM Unit 1117, France; Paris Descartes University, Sorbonne Paris Cité, Division of Infectious Diseases and Tropical Medicine, Necker- Enfants Malades University Hospital, Institut Imagine, Paris, France
| | - Diogo M Magnani
- Department of Pathology, University of Miami, Miami, FL, USA
| | - Caroline Marrs
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| | - Natalia Mercer
- Global Virus Network, 725 West Lombard St., Baltimore, MD, USA
| | | | - Lisa F P Ng
- Global Virus Network, 725 West Lombard St., Baltimore, MD, USA; Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - David H O'Connor
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, USA; Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, USA
| | - Jorge E Osorio
- Global Virus Network, 725 West Lombard St., Baltimore, MD, USA; Department of Pathobiological Sciences, University of Wisconsin-Madison, USA
| | - Guilherme S Ribeiro
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz and Instituto de Saúde Coletiva, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | | | - Shannan L Rossi
- Department of Microbiology & Immunology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - George Saade
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| | - Raymond F Schinazi
- Global Virus Network, 725 West Lombard St., Baltimore, MD, USA; Center for AIDS Research, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Geraldine O Schott-Lerner
- Department of Biochemistry and Molecular Biology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Chao Shan
- Department of Biochemistry and Molecular Biology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - David I Watkins
- Global Virus Network, 725 West Lombard St., Baltimore, MD, USA; Department of Pathology, University of Miami, Miami, FL, USA
| | - Nikos Vasilakis
- Department of Pathology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Scott C Weaver
- Global Virus Network, 725 West Lombard St., Baltimore, MD, USA; Department of Microbiology & Immunology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
171
|
Oh Y, Zhang F, Wang Y, Lee EM, Choi IY, Lim H, Mirakhori F, Li R, Huang L, Xu T, Wu H, Li C, Qin CF, Wen Z, Wu QF, Tang H, Xu Z, Jin P, Song H, Ming GL, Lee G. Zika virus directly infects peripheral neurons and induces cell death. Nat Neurosci 2017; 20:1209-1212. [PMID: 28758997 PMCID: PMC5575960 DOI: 10.1038/nn.4612] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 07/01/2017] [Indexed: 12/16/2022]
Abstract
Zika virus (ZIKV)-infection is associated with neurological disorders of both the central and peripheral nervous systems (PNS), yet few studies have directly examined PNS-infection. Here we show that intraperitoneally or intraventricularly-injected ZIKV in the mouse could infect and impact peripheral neurons in vivo. Moreover, ZIKV productively infects stem cell-derived human neural crest cells and peripheral neurons in vitro, leading to increased cell death, transcriptional dysregulation and cell-type specific molecular pathology.
Collapse
Affiliation(s)
- Yohan Oh
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana, USA
| | - Feiran Zhang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Yaqing Wang
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Emily M Lee
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - In Young Choi
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hotae Lim
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Fahimeh Mirakhori
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ronghua Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Luoxiu Huang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Tianlei Xu
- Department of Mathematics and Computer Science, Emory University, Atlanta, Georgia, USA
| | - Hao Wu
- Department of Biostatistics and Bioinformatics, Emory University Rollins School of Public Health, Atlanta, Georgia, USA
| | - Cui Li
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Cheng-Feng Qin
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Zhexing Wen
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Departments of Psychiatry and Behavioral Sciences, Cell Biology and Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Qing-Feng Wu
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Hengli Tang
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Zhiheng Xu
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,Parkinson's Disease Center, Beijing Institute for Brain Disorders, Beijing, China
| | - Peng Jin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Hongjun Song
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Neuroscience, and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Institute for Regenerative Medicine, Perelman School for Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,The Epigenetics Institute, Perelman School for Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Guo-Li Ming
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Neuroscience, and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Institute for Regenerative Medicine, Perelman School for Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Psychiatry and Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Gabsang Lee
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
172
|
Zika virus preferentially replicates in the female reproductive tract after vaginal inoculation of rhesus macaques. PLoS Pathog 2017; 13:e1006537. [PMID: 28746373 PMCID: PMC5546709 DOI: 10.1371/journal.ppat.1006537] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 08/07/2017] [Accepted: 07/17/2017] [Indexed: 01/18/2023] Open
Abstract
Zika virus (ZIKV) is a mosquito-transmitted virus that can cause severe defects in an infected fetus. ZIKV is also transmitted by sexual contact, although the relative importance of sexual transmission is unclear. To better understand the role of sexual transmission in ZIKV pathogenesis, a nonhuman primate (NHP) model of vaginal transmission was developed. ZIKV was readily transmitted to mature cycling female rhesus macaque (RM) by vaginal inoculation with 104–106 plaque-forming units (PFU). However, there was variability in susceptibility between the individual RM with 1–>8 vaginal inoculations required to establish infection. After treatment with Depoprovera, a widely used contraceptive progestin, two RM that initially resisted 8 vaginal ZIKV inoculations became infected after one ZIKV inoculation. Thus, Depoprovera seemed to enhance susceptibility to vaginal ZIKV transmission. Unexpectedly, the kinetics of virus replication and dissemination after intravaginal ZIKV inoculation were markedly different from RM infected with ZIKV by subcutaneous (SQ) virus inoculation. Several groups have reported that after SQ ZIKV inoculation vRNA is rapidly detected in blood plasma with vRNA less common in urine and saliva and only rarely detected in female reproductive tract (FRT) secretions. In contrast, in vaginally inoculated RM, plasma vRNA is delayed for several days and ZIKV replication in, and vRNA shedding from, the FRT was found in all 6 animals. Further, after intravaginal transmission ZIKV RNA shedding from FRT secretions was detected before or simultaneously with plasma vRNA, and persisted for at least as long. Thus, ZIKV replication in the FRT was independent of, and often preceded virus replication in the tissues contributing to plasma vRNA. These results support the conclusion that ZIKV preferentially replicates in the FRT after vaginal transmission, but not after SQ transmission, and raise the possibility that there is enhanced fetal infection and pathology after vaginal ZIKV transmission compared to a mosquito transmitted ZIKV. Zika virus was introduced to Brazil in 2015 and it rapidly spread to all of tropical America. Although Zika virus infection is usually mild in adults, it can cause severe birth defects in the developing fetus that makes it critical to prevent ZIKV infection in women who are pregnant or who could become pregnant. Although Zika virus is transmitted primarily by mosquito bite, it can also be transmitted by sex. To understand the role of sexual transmission in Zika virus disease, we inoculated rhesus monkeys intravaginally with the virus and monitored virus in blood and reproductive tract secretions. ZIKV was detected in the female reproductive tract before it was detected in plasma and replication levels in the female reproductive tract did not reflect ZIKV levels in other parts of the body. Thus ZIKV prefers the reproductive tract after vaginal transmission suggesting that fetal disease could be more common or severe after vaginal ZIKV transmission compared to a mosquito transmitted ZIKV infection.
Collapse
|
173
|
Zika Virus Persistently Infects and Is Basolaterally Released from Primary Human Brain Microvascular Endothelial Cells. mBio 2017; 8:mBio.00952-17. [PMID: 28698279 PMCID: PMC5513708 DOI: 10.1128/mbio.00952-17] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne Flavivirus that has emerged as the cause of encephalitis and fetal microencephaly in the Americas. ZIKV uniquely persists in human bodily fluids for up to 6 months, is sexually transmitted, and traverses the placenta and the blood-brain barrier (BBB) to damage neurons. Cells that support persistent ZIKV replication and mechanisms by which ZIKV establishes persistence remain enigmatic but central to ZIKV entry into protected neuronal compartments. The endothelial cell (EC) lining of capillaries normally constrains transplacental transmission and forms the BBB, which selectively restricts access of blood constituents to neurons. We found that ZIKV (strain PRVABC59) persistently infects and continuously replicates in primary human brain microvascular ECs (hBMECs), without cytopathology, for >9 days and following hBMEC passage. ZIKV did not permeabilize hBMECs but was released basolaterally from polarized hBMECs, suggesting a direct mechanism for ZIKV to cross the BBB. ZIKV-infected hBMECs were rapidly resistant to alpha interferon (IFN-α) and transiently induced, but failed to secrete, IFN-β and IFN-λ. Global transcriptome analysis determined that ZIKV constitutively induced IFN regulatory factor 7 (IRF7), IRF9, and IFN-stimulated genes (ISGs) 1 to 9 days postinfection, despite persistently replicating in hBMECs. ZIKV constitutively induced ISG15, HERC5, and USP18, which are linked to hepatitis C virus (HCV) persistence and IFN regulation, chemokine CCL5, which is associated with immunopathogenesis, as well as cell survival factors. Our results reveal that hBMECs act as a reservoir of persistent ZIKV replication, suggest routes for ZIKV to cross hBMECs into neuronal compartments, and define novel mechanisms of ZIKV persistence that can be targeted to restrict ZIKV spread.IMPORTANCE ZIKV persists in patients, crossing placental and neuronal barriers, damaging neurons, and causing fetal microencephaly. We found that ZIKV persistently infects brain endothelial cells that normally protect neurons from viral exposure. hBMECs are not damaged by ZIKV infection and, analogous to persistent HCV infection, ZIKV constitutively induces and evades antiviral ISG and IFN responses to continuously replicate in hBMECs. As a result, hBMECs provide a protective niche for systemic ZIKV spread and a viral reservoir localized in the normally protective blood-brain barrier. Consistent with the spread of ZIKV into neuronal compartments, ZIKV was released basolaterally from hBMECs. Our findings define hBMEC responses that contribute to persistent ZIKV infection and potential targets for clearing ZIKV infections from hBMECs. These results further suggest roles for additional ZIKV-infected ECs to facilitate viral spread and persistence in the protected placental, retinal, and testicular compartments.
Collapse
|
174
|
Magalhaes T, Foy BD, Marques ETA, Ebel GD, Weger-Lucarelli J. Mosquito-borne and sexual transmission of Zika virus: Recent developments and future directions. Virus Res 2017; 254:1-9. [PMID: 28705681 DOI: 10.1016/j.virusres.2017.07.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/08/2017] [Accepted: 07/08/2017] [Indexed: 02/04/2023]
Abstract
Zika virus (ZIKV; Genus Flavivirus, Family Flaviviridae) has recently emerged in Asia and the Americas to cause large outbreaks of human disease. The outbreak has been characterized by high attack rates, birth defects in infants and severe neurological complications in adults. ZIKV is transmitted to humans by Aedes mosquitoes, but recent evidence implicates sexual transmission as playing an important role as well. This review highlights the transmission of ZIKV in humans, with a focus on both mosquito and sexually-transmitted routes and their outcomes. We also discuss critical directions for future research.
Collapse
Affiliation(s)
- Tereza Magalhaes
- Department of Microbiology, Immunology and Pathology, Arthropod-borne and Infectious Diseases Laboratory, Colorado State University, Fort Collins, CO, United States
| | - Brian D Foy
- Department of Microbiology, Immunology and Pathology, Arthropod-borne and Infectious Diseases Laboratory, Colorado State University, Fort Collins, CO, United States.
| | - Ernesto T A Marques
- Laboratory of Virology and Experimental Therapeutics, Centro de Pesquisas Aggeu Magalhaes, Fundacao Oswaldo Cruz, Recife, PE, Brazil; Center for Vaccine Research, Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Gregory D Ebel
- Department of Microbiology, Immunology and Pathology, Arthropod-borne and Infectious Diseases Laboratory, Colorado State University, Fort Collins, CO, United States
| | - James Weger-Lucarelli
- Department of Microbiology, Immunology and Pathology, Arthropod-borne and Infectious Diseases Laboratory, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
175
|
Li Z, Brecher M, Deng YQ, Zhang J, Sakamuru S, Liu B, Huang R, Koetzner CA, Allen CA, Jones SA, Chen H, Zhang NN, Tian M, Gao F, Lin Q, Banavali N, Zhou J, Boles N, Xia M, Kramer LD, Qin CF, Li H. Existing drugs as broad-spectrum and potent inhibitors for Zika virus by targeting NS2B-NS3 interaction. Cell Res 2017; 27:1046-1064. [PMID: 28685770 DOI: 10.1038/cr.2017.88] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 05/19/2017] [Accepted: 06/05/2017] [Indexed: 02/07/2023] Open
Abstract
Recent outbreaks of Zika virus (ZIKV) highlight an urgent need for therapeutics. The protease complex NS2B-NS3 plays essential roles during flaviviral polyprotein processing, and thus represents an attractive drug target. Here, we developed a split luciferase complementation-based high-throughput screening assay to identify orthosteric inhibitors that directly target flavivirus NS2B-NS3 interactions. By screening a total of 2 816 approved and investigational drugs, we identified three potent candidates, temoporfin, niclosamide, and nitazoxanide, as flavivirus NS2B-NS3 interaction inhibitors with nanomolar potencies. Significantly, the most potent compound, temoporfin, not only inhibited ZIKV replication in human placental and neural progenitor cells, but also prevented ZIKV-induced viremia and mortality in mouse models. Structural docking suggests that temoporfin potentially binds NS3 pockets that hold critical NS2B residues, thus inhibiting flaviviral polyprotein processing in a non-competitive manner. As these drugs have already been approved for clinical use in other indications either in the USA or other countries, they represent promising and easily developed therapies for the management of infections by ZIKV and other flaviviruses.
Collapse
Affiliation(s)
- Zhong Li
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA
| | - Matthew Brecher
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA
| | - Yong-Qiang Deng
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Jing Zhang
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA
| | - Srilatha Sakamuru
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Binbin Liu
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA.,Department of Food Science, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, Guangdong 524000, China
| | - Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cheri A Koetzner
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA
| | - Christina A Allen
- The Neural Stem Cell Institute, 1 Discovery Drive, Rensselaer, NY 12144, USA
| | - Susan A Jones
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA
| | - Haiying Chen
- Department of Pharmacology and Toxicology, Chemical Biology Program, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Na-Na Zhang
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Min Tian
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Fengshan Gao
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA.,Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Dalian University, Dalian, Liaoning 116622, China
| | - Qishan Lin
- Center for Functional Genomics, University at Albany, Rensselaer, NY 12144, USA
| | - Nilesh Banavali
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA.,Department of Biomedical Sciences, School of Public Health, University at Albany, PO Box 509, Empire State Plaza, Albany, NY 12201, USA
| | - Jia Zhou
- Department of Pharmacology and Toxicology, Chemical Biology Program, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Nathan Boles
- The Neural Stem Cell Institute, 1 Discovery Drive, Rensselaer, NY 12144, USA
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Laura D Kramer
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA.,Department of Biomedical Sciences, School of Public Health, University at Albany, PO Box 509, Empire State Plaza, Albany, NY 12201, USA
| | - Cheng-Feng Qin
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China.,Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Hongmin Li
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA.,Department of Biomedical Sciences, School of Public Health, University at Albany, PO Box 509, Empire State Plaza, Albany, NY 12201, USA
| |
Collapse
|
176
|
Aid M, Abbink P, Larocca RA, Boyd M, Nityanandam R, Nanayakkara O, Martinot AJ, Moseley ET, Blass E, Borducchi EN, Chandrashekar A, Brinkman AL, Molloy K, Jetton D, Tartaglia LJ, Liu J, Best K, Perelson AS, De La Barrera RA, Lewis MG, Barouch DH. Zika Virus Persistence in the Central Nervous System and Lymph Nodes of Rhesus Monkeys. Cell 2017; 169:610-620.e14. [PMID: 28457610 PMCID: PMC5426912 DOI: 10.1016/j.cell.2017.04.008] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 03/12/2017] [Accepted: 04/06/2017] [Indexed: 02/03/2023]
Abstract
Zika virus (ZIKV) is associated with severe neuropathology in neonates as well as Guillain-Barré syndrome and other neurologic disorders in adults. Prolonged viral shedding has been reported in semen, suggesting the presence of anatomic viral reservoirs. Here we show that ZIKV can persist in cerebrospinal fluid (CSF) and lymph nodes (LN) of infected rhesus monkeys for weeks after virus has been cleared from peripheral blood, urine, and mucosal secretions. ZIKV-specific neutralizing antibodies correlated with rapid clearance of virus in peripheral blood but remained undetectable in CSF for the duration of the study. Viral persistence in both CSF and LN correlated with upregulation of mechanistic target of rapamycin (mTOR), proinflammatory, and anti-apoptotic signaling pathways, as well as downregulation of extracellular matrix signaling pathways. These data raise the possibility that persistent or occult neurologic and lymphoid disease may occur following clearance of peripheral virus in ZIKV-infected individuals.
Collapse
Affiliation(s)
- Malika Aid
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Peter Abbink
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Rafael A Larocca
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Michael Boyd
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ramya Nityanandam
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ovini Nanayakkara
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Amanda J Martinot
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Edward T Moseley
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Eryn Blass
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Erica N Borducchi
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Abishek Chandrashekar
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Amanda L Brinkman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Katherine Molloy
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - David Jetton
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Lawrence J Tartaglia
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jinyan Liu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Katharine Best
- Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | | | | | | | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
177
|
Hirsch AJ, Smith JL, Haese NN, Broeckel RM, Parkins CJ, Kreklywich C, DeFilippis VR, Denton M, Smith PP, Messer WB, Colgin LMA, Ducore RM, Grigsby PL, Hennebold JD, Swanson T, Legasse AW, Axthelm MK, MacAllister R, Wiley CA, Nelson JA, Streblow DN. Correction: Zika Virus infection of rhesus macaques leads to viral persistence in multiple tissues. PLoS Pathog 2017; 13:e1006317. [PMID: 28380070 PMCID: PMC5381941 DOI: 10.1371/journal.ppat.1006317] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|