1
|
Imagawa T, Tanaka K, Ito M, Matsuda M, Suzuki T, Ando T, Yaguchi C, Miyamoto K, Takabayashi S, Suzuki R, Takasaki T, Itoh H, Kosugi I, Suzuki T. Pathological characterization of female reproductive organs prior to miscarriage induced by Zika virus infection in the pregnant common marmoset. Microbiol Spectr 2025; 13:e0228224. [PMID: 39998269 PMCID: PMC11960083 DOI: 10.1128/spectrum.02282-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
While Zika virus (ZIKV) infection in pregnant women is known to increase the risk of miscarriage and stillbirth, the mechanism by which ZIKV infection leads to the inability to continue a pregnancy is not clear. In our common marmoset models of ZIKV infection in pregnant individuals, miscarriage was observed in dams infected in the first or second trimester, and preterm delivery was observed in a dam infected in the third trimester. Serum progesterone levels were significantly lower prior to miscarriage or preterm delivery in the infected marmosets. To elucidate the pathology of the placental region just before the onset of ZIKV-induced miscarriage, we newly prepared an infected marmoset in the first trimester of pregnancy and euthanized it when the serum progesterone concentration was markedly reduced. Pathological analysis revealed significant degeneration in cells at the maternal-fetal interface, presumably trophoblasts. Cleaved-caspase was widely observed in the endometrial to placental region, and TNFα at 200 pg/mL was detected in the amniotic fluid, suggesting that apoptosis may progress in the endometrium and placenta, leading to decreased trophoblast function and miscarriage. ZIKV NS1 protein was found sporadically in the cellular degeneration area and widely in the basal layer of the endometrium. Furthermore, the viral protein was frequently detected in the follicles and corpus luteum of the ovary. The developed ZIKV infection model in pregnant marmosets would be useful not only to better understand the mechanism of ZIKV-induced miscarriage but also to analyze the effects of the viral infection on female reproductive tissues. IMPORTANCE Although several viruses, including Zika virus (ZIKV), are known to increase the risk of miscarriage upon viral infection, the mechanism by which miscarriage is induced by viral infection is largely unknown. This is partly due to the difficulty of pathological analysis of maternal tissues in the period following viral infection and prior to miscarriage. In this study, we predicted the occurrence of miscarriage by monitoring serum progesterone levels and performed pathological analysis of peri-placental tissues at a time point assumed to be just before miscarriage. This is the first report of trophoblast degeneration prior to miscarriage, suggesting that the experimental method used here is useful for analyzing the pathogenesis of virus infection-related miscarriage. Further immunostaining revealed that ZIKV NS1 was distributed not only in the uterus but also in the ovaries, with particularly pronounced staining of oocytes. Whether ZIKV infection affects female reproductive function should be clarified in the future.
Collapse
Affiliation(s)
- Toshifumi Imagawa
- Department of Medical Virology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- Department of Microbiology and Immunology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuo Tanaka
- Laboratory Animal Facilities and Services, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masahiko Ito
- Department of Microbiology and Immunology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Mami Matsuda
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tsuyoshi Ando
- Research and Development Division, FUJIREBIO INC., Tokyo, Japan
| | - Chizuko Yaguchi
- Department of Obstetrics and Gynecology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | | | - Shuji Takabayashi
- Laboratory Animal Facilities and Services, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Ryosuke Suzuki
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tomohiko Takasaki
- Advanced Technology and Development Division, BML, INC., Kawagoe, Japan
| | - Hiroaki Itoh
- Department of Obstetrics and Gynecology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Isao Kosugi
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tetsuro Suzuki
- Department of Microbiology and Immunology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
2
|
Li A, Coffey LL, Mohr EL, Raper J, Chahroudi A, Ausderau KK, Aliota MT, Friedrich TC, Mitzey AM, Koenig MR, Golos TG, Jaeger HK, Roberts VHJ, Lo JO, Smith JL, Hirsch AJ, Streblow DN, Newman CM, O'Connor DH, Lackritz EM, Van Rompay KKA, Adams Waldorf KM. Role of non-human primate models in accelerating research and developing countermeasures against Zika virus infection. THE LANCET. MICROBE 2025:101030. [PMID: 40024258 DOI: 10.1016/j.lanmic.2024.101030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/19/2024] [Accepted: 10/21/2024] [Indexed: 03/04/2025]
Abstract
Zika virus, a mosquito-transmitted orthoflavivirus, has become a pathogen of global health concern ever since the virus caused an epidemic in Brazil in 2015 associated with approximately 700 000 laboratory-confirmed cases of congenital microcephaly. The subsequent spread of the epidemic in 2016 resulted in a wide spectrum of congenital neurological, ophthalmological, and developmental abnormalities across the Americas, Africa, and Asia. In this context, non-human primate models have become essential tools for Zika virus research to understand the pathogenesis of congenital brain injury and perinatal complications and for developing and testing medical countermeasures such as vaccines, diagnostics, and therapeutics. Fetal brain injury has been observed across various non-human primate species and is influenced by factors such as the Zika virus strain, gestational age at inoculation, and inoculation dose and route. Miscarriages are also seen as common outcomes of first trimester Zika virus infections. This Series paper reviews the diverse non-human primate models currently used for Zika virus research to mitigate the public health effects of future Zika virus epidemics.
Collapse
Affiliation(s)
- Amanda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Lark L Coffey
- Department of Pathology, Microbiology, and Immunology, University of California, Davis, Davis, CA, USA
| | - Emma L Mohr
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA; Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Jessica Raper
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA; Emory National Primate Research Center, Atlanta, GA, USA
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA; Emory National Primate Research Center, Atlanta, GA, USA
| | - Karla K Ausderau
- Department of Kinesiology, University of Wisconsin-Madison, Madison, WI, USA; Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Matthew T Aliota
- Department of Veterinary and Biomedical Sciences, University of Minnesota Twin Cities, St Paul, MN, USA
| | - Thomas C Friedrich
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA; Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Ann M Mitzey
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelle R Koenig
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Thaddeus G Golos
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA; Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Hannah K Jaeger
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, USA
| | - Victoria H J Roberts
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Jamie O Lo
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR, USA; Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Jessica L Smith
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, USA
| | - Alec J Hirsch
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, USA
| | - Daniel N Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, USA
| | - Christina M Newman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - David H O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA; Wisconsin National Primate Research Center, Madison, WI, USA
| | - Eve M Lackritz
- Center for Infectious Disease Research and Policy (CIDRAP), University of Minnesota, Minneapolis, MN, USA
| | - Koen K A Van Rompay
- Department of Pathology, Microbiology, and Immunology, University of California, Davis, Davis, CA, USA; California National Primate Research Center, Davis, CA, USA
| | - Kristina M Adams Waldorf
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA; Washington National Primate Research Center, Seattle, WA, USA.
| |
Collapse
|
3
|
Wang J, Yu Z, Chen Z, Ye F, Sun Z. The Potential Role of Zika and Dengue Virus Infection in the Urogenital System Disorders: An Overview. Rev Med Virol 2025; 35:e70010. [PMID: 39804234 DOI: 10.1002/rmv.70010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 05/02/2025]
Abstract
Arboviruses currently are regarded as a major worldwide public health concern. The clinical outcomes associated with this group of viruses may vary from asymptomatic infections to severe forms of haemorrhagic fever characterised by bleeding disorders. Similar to other systemic viral infections, arboviruses can either directly or indirectly affect different parts of the body, such as the urogenital system. The human urogenital system anatomically consists of two major subdivisions: (i) the urinary system, including the kidneys, ureters, bladder, and urethra, which plays a significant role in osmoregulation, control of blood volume, pressure, and PH, absorption/excretion of different ions, and toxin metabolism, and (ii) the genital system, composed of the prostate, uterus, testes, ovaries, penis, and vagina, which are responsible for reproductive functions. Arboviruses can impair normal urogenital system functions by direct viral pathogen activity, systemic forms of inflammation, haemorrhagic events and related dysfunctions, and the nephrotoxic side effects of specific medications employed for treatment leading to various urogenital disorders. The present review provides an overview of the potential capacity of two main arboviruses, known as Zika and dengue viruses, to affect the urogenital system. Moreover, it addresses Zika virus as a potential therapeutic oncolytic virus for urogenital cancers.
Collapse
Affiliation(s)
- Jie Wang
- Department of Urology, The Second People's Hospital of Meishan City, Sichuan, China
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zongze Yu
- Department of Urology, The Second People's Hospital of Meishan City, Sichuan, China
| | - Zhigui Chen
- Department of Urology, The Second People's Hospital of Meishan City, Sichuan, China
| | - Fangdie Ye
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhou Sun
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
4
|
Shofa M, Okamura T, Urano E, Matsuura Y, Yasutomi Y, Saito A. Repeated Intravaginal Inoculation of Zika Virus Protects Cynomolgus Monkeys from Subcutaneous Superchallenge. Int J Mol Sci 2022; 23:ijms232214002. [PMID: 36430481 PMCID: PMC9696507 DOI: 10.3390/ijms232214002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/08/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Zika virus (ZIKV) outbreaks in Central and South America caused severe public health problems in 2015 and 2016. These outbreaks were finally contained through several methods, including mosquito control using insecticides and repellents. Additionally, the development of herd immunity in these countries might have contributed to containing the epidemic. While ZIKV is mainly transmitted by mosquito bites and mucosal transmission via bodily fluids, including the semen of infected individuals, has also been reported. We evaluated the effect of mucosal ZIKV infection on continuous subcutaneous challenges in a cynomolgus monkey model. Repeated intravaginal inoculations of ZIKV did not induce detectable viremia or clinical symptoms, and all animals developed a potent neutralizing antibody, protecting animals from the subsequent subcutaneous superchallenge. These results suggest that viral replication at mucosal sites can induce protective immunity without causing systemic viremia or symptoms.
Collapse
Affiliation(s)
- Maya Shofa
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan
- Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Tomotaka Okamura
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki 305-0843, Japan
| | - Emiko Urano
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki 305-0843, Japan
| | - Yoshiharu Matsuura
- Center for Infectious Disease Education and Research, Osaka University, Osaka 565-0871, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Yasuhiro Yasutomi
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki 305-0843, Japan
- Department of Molecular and Experimental Medicine, Mie University Graduate School of Medicine, Mie 514-8507, Japan
- Correspondence: (Y.Y.); (A.S.)
| | - Akatsuki Saito
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan
- Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
- Center for Animal Disease Control, University of Miyazaki, Miyazaki 889-2192, Japan
- Correspondence: (Y.Y.); (A.S.)
| |
Collapse
|
5
|
McMillen CM, Boyles DA, Kostadinov SG, Hoehl RM, Schwarz MM, Albe JR, Demers MJ, Hartman AL. Congenital Rift Valley fever in Sprague Dawley rats is associated with diffuse infection and pathology of the placenta. PLoS Negl Trop Dis 2022; 16:e0010898. [PMID: 36315601 PMCID: PMC9648853 DOI: 10.1371/journal.pntd.0010898] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 11/10/2022] [Accepted: 10/17/2022] [Indexed: 11/12/2022] Open
Abstract
Rift Valley fever (RVF) is a disease of animals and humans associated with abortions in ruminants and late-gestation miscarriages in women. Here, we use a rat model of congenital RVF to identify tropisms, pathologies, and immune responses in the placenta during vertical transmission. Infection of late-gestation pregnant rats resulted in vertical transmission to the placenta and widespread infection throughout the decidua, basal zone, and labyrinth zone. Some pups from infected dams appeared normal while others had gross signs of teratogenicity including death. Histopathological lesions were detected in placenta from pups regardless of teratogenicity, while teratogenic pups had widespread hemorrhage throughout multiple placenta layers. Teratogenic events were associated with significant increases in placental pro-inflammatory cytokines, type I interferons, and chemokines. RVFV displays a high degree of tropism for all placental tissue layers and the degree of hemorrhage and inflammatory mediator production is highest in placenta from pups with adverse outcomes. Given the potential for RVFV to emerge in new locations and the recent evidence of emerging viruses, like Zika and SARS-CoV-2, to undergo vertical transmission, this study provides essential understanding regarding the mechanisms by which RVFV crosses the placenta barrier. Rift Valley fever virus (RVFV) infections cause human health and economical burdens given its ability to induce high rates of abortions in ruminants and possible contributions towards late-term miscarriages in women. In this study, we have identified important structures in the placenta targeted by this emerging bunyavirus. Inflammation was associated with more severe fetal outcomes such as death and fetal deformities. The striking similarities between the pathologies of the placenta in the rat model of congenital RVF and those observed in naturally infected ruminants highlight the utility of this rodent model. These findings may be further translated towards understanding the mechanisms involved in vertical transmission of RVFV in humans.
Collapse
Affiliation(s)
- Cynthia M. McMillen
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Devin A. Boyles
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Stefan G. Kostadinov
- Department of Pathology, Magee Women’s Hospital of the University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - Ryan M. Hoehl
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Madeline M. Schwarz
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Joseph R. Albe
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Matthew J. Demers
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Amy L. Hartman
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
6
|
Kim S, Shin HY. Understanding the Tissue Specificity of ZIKV Infection in Various Animal Models for Vaccine Development. Vaccines (Basel) 2022; 10:1517. [PMID: 36146595 PMCID: PMC9504629 DOI: 10.3390/vaccines10091517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/08/2022] [Accepted: 09/10/2022] [Indexed: 11/23/2022] Open
Abstract
Zika virus (ZIKV) is an arthropod-borne virus that belongs to the Flavivirus genus and is principally transmitted by Aedes aegypti mosquitoes. ZIKV infection often causes no or only mild symptoms, but it can also trigger severe consequences, including microcephaly in infants and Guillain-Barré syndrome, uveitis, and neurologic manifestations in adults. There is no ZIKV vaccine or treatment currently approved for clinical use. The primary target of ZIKV infection has been recognized as the maternal placenta, with vertical transmission to the fetal brain. However, ZIKV can also spread to multiple tissues in adults, including the sexual organs, eyes, lymph nodes, and brain. Since numerous studies have indicated that there are slightly different tissue-specific pathologies in each animal model of ZIKV, the distinct ZIKV tropism of a given animal model must be understood to enable effective vaccine development. Here, we comprehensively discussed the tissue specificity of ZIKV reported in each animal model depending on the genetic background and route of administration. This review should facilitate the selection of appropriate animal models when studying the fundamental pathogenesis of ZIKV infection, thereby supporting the design of optimal preclinical and clinical studies for the development of vaccines and therapeutics.
Collapse
Affiliation(s)
| | - Ha Youn Shin
- Department of Biomedical Science and Engineering, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
7
|
Berry N, Stein M, Ferguson D, Ham C, Hall J, Giles E, Kempster S, Adedeji Y, Almond N, Herrera C. Mucosal Responses to Zika Virus Infection in Cynomolgus Macaques. Pathogens 2022; 11:1033. [PMID: 36145466 PMCID: PMC9503824 DOI: 10.3390/pathogens11091033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 12/03/2022] Open
Abstract
Zika virus (ZIKV) cases continue to be reported, and no vaccine or specific antiviral agent has been approved for the prevention or treatment of infection. Though ZIKV is primarily transmitted by mosquitos, cases of sexual transmission and prolonged viral RNA presence in semen have been reported. In this observational study, we report the mucosal responses to sub-cutaneous and mucosal ZIKV exposure in cynomolgus macaques during acute and late chronic infection. Subcutaneous challenge induced a decrease in the growth factor VEGF in colorectal and cervicovaginal tissues 100 days post-challenge, in contrast to the observed increase in these tissues following vaginal infection. This different pattern was not observed in the uterus, where VEGF was upregulated independently of the challenge route. Vaginal challenge induced a pro-inflammatory profile in all mucosal tissues during late chronic infection. Similar responses were already observed during acute infection in a vaginal tissue explant model of ex vivo challenge. Non-productive and productive infection 100 days post-in vivo vaginal challenge induced distinct proteomic profiles which were characterized by further VEGF increase and IL-10 decrease in non-infected animals. Ex vivo challenge of mucosal explants revealed tissue-specific modulation of cytokine levels during the acute phase of infection. Mucosal cytokine profiles could represent biosignatures of persistent ZIKV infection.
Collapse
Affiliation(s)
- Neil Berry
- Division of Infectious Disease Diagnostics, National Institute for Biological Standards and Control (NIBSC), Potters Bar EN6 3QC, UK
| | - Monja Stein
- Department of Medicine, Imperial College London, London W2 1PG, UK
| | - Deborah Ferguson
- Division of Infectious Disease Diagnostics, National Institute for Biological Standards and Control (NIBSC), Potters Bar EN6 3QC, UK
| | - Claire Ham
- Division of Infectious Disease Diagnostics, National Institute for Biological Standards and Control (NIBSC), Potters Bar EN6 3QC, UK
| | - Jo Hall
- Division of Infectious Disease Diagnostics, National Institute for Biological Standards and Control (NIBSC), Potters Bar EN6 3QC, UK
| | - Elaine Giles
- Division of Analytical and Biological Sciences, NIBSC, Potters Bar EN6 3QC, UK
| | - Sarah Kempster
- Division of Infectious Disease Diagnostics, National Institute for Biological Standards and Control (NIBSC), Potters Bar EN6 3QC, UK
| | - Yemisi Adedeji
- Division of Infectious Disease Diagnostics, National Institute for Biological Standards and Control (NIBSC), Potters Bar EN6 3QC, UK
| | - Neil Almond
- Division of Infectious Disease Diagnostics, National Institute for Biological Standards and Control (NIBSC), Potters Bar EN6 3QC, UK
| | - Carolina Herrera
- Department of Medicine, Imperial College London, London W2 1PG, UK
| |
Collapse
|
8
|
Mungin JW, Chen X, Liu B. Interferon Epsilon Signaling Confers Attenuated Zika Replication in Human Vaginal Epithelial Cells. Pathogens 2022; 11:853. [PMID: 36014974 PMCID: PMC9415962 DOI: 10.3390/pathogens11080853] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/18/2022] [Accepted: 07/27/2022] [Indexed: 02/07/2023] Open
Abstract
Zika virus (ZIKV) is an emerging flavivirus that causes congenital birth defects and neurological compilations in the human host. Although ZIKV is primarily transmitted through infected mosquitos, recent studies reveal sexual contact as a potential transmission route. In vagina-bearing individuals, the vaginal epithelium constitutes the first line of defense against viruses. However, it is unclear how ZIKV interacts with the vaginal epithelium to initiate ZIKV transmission. In this study, we demonstrate that exposing ZIKV to human vaginal epithelial cells (hVECs) resulted in de novo viral RNA replication, increased envelope viral protein production, and a steady, extracellular release of infectious viral particles. Interestingly, our data show that, despite an increase in viral load, the hVECs did not exhibit significant cytopathology in culture as other cell types typically do. Furthermore, our data reveal that the innate antiviral state of hVECs plays a crucial role in preventing viral cytopathology. For the first time, our data show that interferon epsilon inhibits ZIKV replication. Collectively, our results in this study provide a novel perspective on the viral susceptibility and replication dynamics during ZIKV infection in the human vaginal epithelium. These findings will be instrumental towards developing therapeutic agents aimed at eliminating the pathology caused by the virus.
Collapse
Affiliation(s)
| | | | - Bindong Liu
- Centers for AIDS Health Disparity Research, Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN 37208, USA; (J.W.M.J.); (X.C.)
| |
Collapse
|
9
|
Watts JL, Ralston A. The fetal lineage is susceptible to Zika virus infection within days of fertilization. Development 2022; 149:276104. [PMID: 35900100 PMCID: PMC9382896 DOI: 10.1242/dev.200501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 06/09/2022] [Indexed: 11/20/2022]
Abstract
Adults contracting Zika virus (ZIKV) typically exhibit mild symptoms, yet ZIKV infection of pregnant individuals can cause miscarriage or birth defects in their offspring. Many studies have focused on maternal-to-fetal ZIKV transmission via blood and placenta. Notably, however, ZIKV is also transmitted sexually, raising the possibility that ZIKV could infect the embryo shortly after fertilization, long before the placenta is established. Here, we evaluate the consequences of ZIKV infection in mouse embryos during the first few days of embryogenesis. We show that divergent strains of ZIKV can infect the fetal lineage and can cause developmental arrest, raising concern for the developmental consequences of sexual ZIKV transmission. This article has an associated ‘The people behind the papers’ interview. Summary: Mouse preimplantation embryos are vulnerable to Zika virus-induced lethality even in the presence of the zona pellucida, highlighting a potential risk of sexually transmitted infection in early pregnancy.
Collapse
Affiliation(s)
- Jennifer L. Watts
- Molecular, Cellular and Integrative Physiology Graduate Program, Michigan State University 1 , East Lansing , MI 48824 , USA
- Michigan State University 2 Reproductive and Developmental Biology Training Program , , East Lansing , MI 48824 , USA
- Michigan State University 3 Department of Biochemistry and Molecular Biology , , East Lansing , MI 48824 , USA
| | - Amy Ralston
- Michigan State University 2 Reproductive and Developmental Biology Training Program , , East Lansing , MI 48824 , USA
- Michigan State University 3 Department of Biochemistry and Molecular Biology , , East Lansing , MI 48824 , USA
| |
Collapse
|
10
|
Mask E, Hodara VL, Callery JE, Parodi LM, Obregon-Perko V, Yagi S, Glenn J, Frost P, Clemmons E, Patterson JL, Cox LA, Giavedoni LD. Molecular Approaches for the Validation of the Baboon as a Nonhuman Primate Model for the Study of Zika Virus Infection. Front Cell Infect Microbiol 2022; 12:880860. [PMID: 35493734 PMCID: PMC9046911 DOI: 10.3389/fcimb.2022.880860] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Nonhuman primates (NHP) are particularly important for modeling infections with viruses that do not naturally replicate in rodent cells. Zika virus (ZIKV) has been responsible for sporadic epidemics, but in 2015 a disseminated outbreak of ZIKV resulted in the World Health Organization declaring it a global health emergency. Since the advent of this last epidemic, several NHP species, including the baboon, have been utilized for modeling and understanding the complications of ZIKV infection in humans; several health issues related to the outcome of infection have not been resolved yet and require further investigation. This study was designed to validate, in baboons, the molecular signatures that have previously been identified in ZIKV-infected humans and macaque models. We performed a comprehensive molecular analysis of baboons during acute ZIKV infection, including flow cytometry, cytokine, immunological, and transcriptomic analyses. We show here that, similar to most human cases, ZIKV infection of male baboons tends to be subclinical, but is associated with a rapid and transient antiviral interferon-based response signature that induces a detectable humoral and cell-mediated immune response. This immunity against the virus protects animals from challenge with a divergent ZIKV strain, as evidenced by undetectable viremia but clear anamnestic responses. These results provide additional support for the use of baboons as an alternative animal model to macaques and validate omic techniques that could help identify the molecular basis of complications associated with ZIKV infections in humans.
Collapse
Affiliation(s)
- Emma Mask
- Department of Biology, Trinity University, San Antonio, TX, United States
| | - Vida L. Hodara
- Southwest National Primate Research Center, San Antonio, TX, United States,Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Jessica E. Callery
- Department of Biology, Trinity University, San Antonio, TX, United States
| | - Laura M. Parodi
- Southwest National Primate Research Center, San Antonio, TX, United States,Texas Biomedical Research Institute, San Antonio, TX, United States
| | | | - Shigeo Yagi
- California Department of Public Health, Richmond, CA, United States
| | - Jeremy Glenn
- Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Patrice Frost
- Southwest National Primate Research Center, San Antonio, TX, United States
| | - Elizabeth Clemmons
- Southwest National Primate Research Center, San Antonio, TX, United States
| | | | - Laura A. Cox
- Southwest National Primate Research Center, San Antonio, TX, United States,Center for Precision Medicine, Wake Forest Health Sciences University, Winston Salem, NC, United States
| | - Luis D. Giavedoni
- Department of Biology, Trinity University, San Antonio, TX, United States,Southwest National Primate Research Center, San Antonio, TX, United States,*Correspondence: Luis D. Giavedoni,
| |
Collapse
|
11
|
Are the Organoid Models an Invaluable Contribution to ZIKA Virus Research? Pathogens 2021; 10:pathogens10101233. [PMID: 34684182 PMCID: PMC8537471 DOI: 10.3390/pathogens10101233] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/15/2021] [Accepted: 09/20/2021] [Indexed: 12/16/2022] Open
Abstract
In order to prevent new pathogen outbreaks and avoid possible new global health threats, it is important to study the mechanisms of microbial pathogenesis, screen new antiviral agents and test new vaccines using the best methods. In the last decade, organoids have provided a groundbreaking opportunity for modeling pathogen infections in human brains, including Zika virus (ZIKV) infection. ZIKV is a member of the Flavivirus genus, and it is recognized as an emerging infectious agent and a serious threat to global health. Organoids are 3D complex cellular models that offer an in-scale organ that is physiologically alike to the original one, useful for exploring the mechanisms behind pathogens infection; additionally, organoids integrate data generated in vitro with traditional tools and often support those obtained in vivo with animal model. In this mini-review the value of organoids for ZIKV research is examined and sustained by the most recent literature. Within a 3D viewpoint, tissue engineered models are proposed as future biological systems to help in deciphering pathogenic processes and evaluate preventive and therapeutic strategies against ZIKV. The next steps in this field constitute a challenge that may protect people and future generations from severe brain defects.
Collapse
|
12
|
Newman CM, Tarantal AF, Martinez ML, Simmons HA, Morgan TK, Zeng X, Rosinski JR, Bliss MI, Bohm EK, Dudley DM, Aliota MT, Friedrich TC, Miller CJ, O’Connor DH. Early Embryonic Loss Following Intravaginal Zika Virus Challenge in Rhesus Macaques. Front Immunol 2021; 12:686437. [PMID: 34079560 PMCID: PMC8165274 DOI: 10.3389/fimmu.2021.686437] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/04/2021] [Indexed: 01/25/2023] Open
Abstract
Zika virus (ZIKV) is an arthropod-borne virus (arbovirus) and is primarily transmitted by Aedes species mosquitoes; however, ZIKV can also be sexually transmitted. During the initial epidemic and in places where ZIKV is now considered endemic, it is difficult to disentangle the risks and contributions of sexual versus vector-borne transmission to adverse pregnancy outcomes. To examine the potential impact of sexual transmission of ZIKV on pregnancy outcome, we challenged three rhesus macaques (Macaca mulatta) three times intravaginally with 1 x 107 PFU of a low passage, African lineage ZIKV isolate (ZIKV-DAK) in the first trimester (~30 days gestational age). Samples were collected from all animals initially on days 3 through 10 post challenge, followed by twice, and then once weekly sample collection; ultrasound examinations were performed every 3-4 days then weekly as pregnancies progressed. All three dams had ZIKV RNA detectable in plasma on day 3 post-ZIKV challenge. At approximately 45 days gestation (17-18 days post-challenge), two of the three dams were found with nonviable embryos by ultrasound. Viral RNA was detected in recovered tissues and at the maternal-fetal interface (MFI) in both cases. The remaining viable pregnancy proceeded to near term (~155 days gestational age) and ZIKV RNA was detected at the MFI but not in fetal tissues. These results suggest that sexual transmission of ZIKV may represent an underappreciated risk of pregnancy loss during early gestation.
Collapse
Affiliation(s)
- Christina M. Newman
- Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, United States
| | - Alice F. Tarantal
- Pediatrics, Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, CA, United States
- California National Primate Research Center, University of California, Davis, CA, United States
| | - Michele L. Martinez
- Pediatrics, Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, CA, United States
- California National Primate Research Center, University of California, Davis, CA, United States
| | - Heather A. Simmons
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Terry K. Morgan
- Pathology, Oregon Health and Sciences University, Portland, OR, United States
| | - Xiankun Zeng
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Jenna R. Rosinski
- Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, United States
| | - Mason I. Bliss
- Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Ellie K. Bohm
- Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, MN, United States
| | - Dawn M. Dudley
- Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, United States
| | - Matthew T. Aliota
- Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, MN, United States
| | - Thomas C. Friedrich
- Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Christopher J. Miller
- California National Primate Research Center, University of California, Davis, CA, United States
- Pathology, Microbiology, and Immunology, School of Veterinary Medicine, Center for Immunology and Infectious Diseases, University of California, Davis, CA, United States
| | - David H. O’Connor
- Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, United States
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
13
|
Xiong R, Zhang L, Li S, Sun Y, Ding M, Wang Y, Zhao Y, Wu Y, Shang W, Jiang X, Shan J, Shen Z, Tong Y, Xu L, Chen Y, Liu Y, Zou G, Lavillete D, Zhao Z, Wang R, Zhu L, Xiao G, Lan K, Li H, Xu K. Novel and potent inhibitors targeting DHODH are broad-spectrum antivirals against RNA viruses including newly-emerged coronavirus SARS-CoV-2. Protein Cell 2020; 11:723-739. [PMID: 32754890 DOI: 10.1101/2020.03.11.983056] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 07/05/2020] [Indexed: 05/18/2023] Open
Abstract
Emerging and re-emerging RNA viruses occasionally cause epidemics and pandemics worldwide, such as the on-going outbreak of the novel coronavirus SARS-CoV-2. Herein, we identified two potent inhibitors of human DHODH, S312 and S416, with favorable drug-likeness and pharmacokinetic profiles, which all showed broad-spectrum antiviral effects against various RNA viruses, including influenza A virus, Zika virus, Ebola virus, and particularly against SARS-CoV-2. Notably, S416 is reported to be the most potent inhibitor so far with an EC50 of 17 nmol/L and an SI value of 10,505.88 in infected cells. Our results are the first to validate that DHODH is an attractive host target through high antiviral efficacy in vivo and low virus replication in DHODH knock-out cells. This work demonstrates that both S312/S416 and old drugs (Leflunomide/Teriflunomide) with dual actions of antiviral and immuno-regulation may have clinical potentials to cure SARS-CoV-2 or other RNA viruses circulating worldwide, no matter such viruses are mutated or not.
Collapse
Affiliation(s)
- Rui Xiong
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Leike Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Shiliang Li
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yuan Sun
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Minyi Ding
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yong Wang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Yongliang Zhao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Yan Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Weijuan Shang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xiaming Jiang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jiwei Shan
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Zihao Shen
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yi Tong
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Liuxin Xu
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yu Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Yingle Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Gang Zou
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Dimitri Lavillete
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zhenjiang Zhao
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Rui Wang
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Lili Zhu
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Gengfu Xiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Ke Lan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Honglin Li
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| | - Ke Xu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
14
|
de Souza AAA, Torres LR, Lima LRP, de Paula V, Barros JJ, Bonecini-Almeida MDG, Waghabi MC, Gardel MA, Meuser-Batista M, de Souza EM. Inhibition of Brazilian ZIKV strain replication in primary human placental chorionic cells and cervical cells treated with nitazoxanide. Braz J Infect Dis 2020; 24:505-516. [PMID: 33010209 PMCID: PMC7526660 DOI: 10.1016/j.bjid.2020.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/28/2020] [Accepted: 09/08/2020] [Indexed: 01/26/2023] Open
Abstract
Zika virus (ZIKV) infection during pregnancy is associated with a congenital syndrome. Although the virus can be detected in human placental tissue and sexual transmission has been verified, it is not clear how the virus reaches the fetus. Despite the emerging severity caused by ZIKV infection, no specific prophylactic and/or therapeutic treatment is available. The aim of the present study was to evaluate the effectiveness antiviral of nitazoxanide (NTZ) in two important congenital transmission targets: (i) a primary culture of human placental chorionic cells, and (ii) human cervical epithelial cells (C33-A) infected with Brazilian ZIKV strain. Initially, NTZ activity was screened in ZIKV infected Vero cells under different treatment regimens with non-toxic drug concentrations for 48 h. Antiviral effect was found only when the treatment was carried out after the viral inoculum. A strong effect against the dengue virus serotype 2 (DENV-2) was also observed suggesting the possibility of treating other Flaviviruses. Additionally, it was shown that the treatment did not reduce the production of infectious viruses in insect cells (C6/36) infected with ZIKV, indicating that the activity of this drug is also related to host factors. Importantly, we demonstrated that NTZ treatment in chorionic and cervical cells caused a reduction of infected cells in a dose-dependent manner and decreased viral loads in up to 2 logs. Pre-clinical in vitro testing evidenced excellent therapeutic response of infected chorionic and cervical cells and point to future NTZ activity investigation in ZIKV congenital transmission models with the perspective of possible repurposing of NTZ to treat Zika fever, especially in pregnant women.
Collapse
Affiliation(s)
- Audrien A A de Souza
- Instituto Oswaldo Cruz/FIOCRUZ, Laboratório de Virologia Molecular, Rio de Janeiro, RJ, Brazil
| | - Lauana R Torres
- Instituto Oswaldo Cruz/FIOCRUZ, Laboratório de Virologia Molecular, Rio de Janeiro, RJ, Brazil
| | - Lyana R P Lima
- Instituto Oswaldo Cruz/FIOCRUZ, Laboratório de Virologia Molecular, Rio de Janeiro, RJ, Brazil
| | - Vanessa de Paula
- Instituto Oswaldo Cruz/FIOCRUZ, Laboratório de Virologia Molecular, Rio de Janeiro, RJ, Brazil
| | - José J Barros
- Instituto Oswaldo Cruz/FIOCRUZ, Laboratório de Virologia Molecular, Rio de Janeiro, RJ, Brazil
| | - Maria da Gloria Bonecini-Almeida
- Instituto Nacional de Infectologia Evandro Chagas/FIOCRUZ, Laboratório de Imunologia e Imunogenética em Doenças Infecciosas, Rio de Janeiro, RJ, Brazil
| | - Mariana Caldas Waghabi
- Instituto Oswaldo Cruz/FIOCRUZ, Laboratório de Genômica Funcional e Bioinformática, Rio de Janeiro, RJ, Brazil
| | - Marcelo A Gardel
- Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira/FIOCRUZ, Coordenação Diagnóstica de Anatomia Patológica e Citopatologia, Rio de Janeiro, RJ, Brazil
| | - Marcelo Meuser-Batista
- Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira/FIOCRUZ, Coordenação Diagnóstica de Anatomia Patológica e Citopatologia, Rio de Janeiro, RJ, Brazil
| | - Elen M de Souza
- Instituto Oswaldo Cruz/FIOCRUZ, Laboratório de Virologia Molecular, Rio de Janeiro, RJ, Brazil; Instituto Oswaldo Cruz/FIOCRUZ, Laboratório de Morfologia e Morfogênese Viral, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
15
|
Wang R, Gornalusse GG, Kim Y, Pandey U, Hladik F, Vojtech L. Potent Restriction of Sexual Zika Virus Infection by the Lipid Fraction of Extracellular Vesicles in Semen. Front Microbiol 2020; 11:574054. [PMID: 33133043 PMCID: PMC7550675 DOI: 10.3389/fmicb.2020.574054] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/07/2020] [Indexed: 12/31/2022] Open
Abstract
Sexual Zika virus (ZIKV) transmission from men to women occurs less frequently than the often-detected high viral loads in semen would suggest, but worries that this transmission route predisposes to fetal damage in pregnant women remain. To better understand sexual ZIKV pathogenesis, we studied the permissiveness of the human female genital tract to infection and the effect of semen on this process. ZIKV replicates in vaginal tissues and primary epithelial cells from the vagina, ectocervix, and endocervix and induces an innate immune response, but also continues to replicate without cytopathic effect. Infection of genital cells and tissues is strongly inhibited by extracellular vesicles (EV) in semen at physiological vesicle-to-virus ratios. Liposomes with the same composition as semen EVs also impair infection, indicating that the EV’s lipid fraction, rather than their protein or RNA cargo, is responsible for this anti-viral effect. Thus, EVs in semen potently restrict ZIKV transmission, but the virus propagates well once infection in the recipient mucosa has been established.
Collapse
Affiliation(s)
- Ruofan Wang
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Germán G Gornalusse
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Yeseul Kim
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Urvashi Pandey
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Florian Hladik
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States.,Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA, United States.,Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Lucia Vojtech
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| |
Collapse
|
16
|
Passive immunisation of convalescent human anti-Zika plasma protects against challenge with New World Zika virus in cynomolgus macaques. NPJ Vaccines 2020; 5:86. [PMID: 33014434 PMCID: PMC7492244 DOI: 10.1038/s41541-020-00234-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022] Open
Abstract
Zika virus (ZIKV) causes neurological complications in susceptible individuals, highlighted in the recent South American epidemic. Natural ZIKV infection elicits host responses capable of preventing subsequent re-infection, raising expectations for effective vaccination. Defining protective immune correlates will inform viral intervention strategies, particularly vaccine development. Non-human primate (NHP) species are susceptible to ZIKV and represent models for vaccine development. The protective efficacy of a human anti-ZIKV convalescent plasma pool (16/320-14) developed as a candidate reference material for a WHO International Standard was evaluated in macaques. Convalescent plasma administered to four cynomolgus macaques (Macaca fascicularis) intra-peritoneally 24 hrs prior to sub-cutaneous challenge with 103 pfu ZIKVPRVABC59 protected against detectable infection, with absence of detectable ZIKV RNA in blood and lymphoid tissues. Passively immunised anti-ZIKV immunoglobulin administered prior to time of challenge remained present only at very low levels 42 days post-challenge. Absence of de novo antibody responses in passively immunised macaques indicate sterilising immunity compared with naïve challenge controls that exhibited active ZIKV-specific IgM and IgG responses post-challenge. Demonstration that the presence of convalescent anti-ZIKV at levels of 400 IU/mL neutralising antibody protects against virus challenge provides a scientific framework for development of anti-ZIKV vaccines and facilitates regulatory approval.
Collapse
|
17
|
Blitvich BJ, Magalhaes T, Laredo-Tiscareño SV, Foy BD. Sexual Transmission of Arboviruses: A Systematic Review. Viruses 2020; 12:v12090933. [PMID: 32854298 PMCID: PMC7552039 DOI: 10.3390/v12090933] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 12/15/2022] Open
Abstract
Arthropod-borne viruses (arboviruses) are primarily maintained in nature in transmission cycles between hematophagous arthropods and vertebrate hosts, but an increasing number of arboviruses have been isolated from or indirectly detected in the urogenital tract and sexual secretions of their vertebrate hosts, indicating that further investigation on the possibility of sexual transmission of these viruses is warranted. The most widely recognized sexually-transmitted arbovirus is Zika virus but other arboviruses, including Crimean-Congo hemorrhagic fever virus and dengue virus, might also be transmitted, albeit occasionally, by this route. This review summarizes our current understanding on the ability of arboviruses to be sexually transmitted. We discuss the sexual transmission of arboviruses between humans and between vertebrate animals, but not arthropod vectors. Every taxonomic group known to contain arboviruses (Asfarviridae, Bunyavirales, Flaviviridae, Orthomyxoviridae, Reoviridae, Rhabdoviridae and Togaviridae) is covered.
Collapse
Affiliation(s)
- Bradley J. Blitvich
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA;
- Correspondence: ; Tel.: +1-515-294-9861; Fax: +1-515-294-8500
| | - Tereza Magalhaes
- Arthropod-Borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (T.M.); (B.D.F.)
| | - S. Viridiana Laredo-Tiscareño
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA;
| | - Brian D. Foy
- Arthropod-Borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (T.M.); (B.D.F.)
| |
Collapse
|
18
|
Xiong R, Zhang L, Li S, Sun Y, Ding M, Wang Y, Zhao Y, Wu Y, Shang W, Jiang X, Shan J, Shen Z, Tong Y, Xu L, Chen Y, Liu Y, Zou G, Lavillete D, Zhao Z, Wang R, Zhu L, Xiao G, Lan K, Li H, Xu K. Novel and potent inhibitors targeting DHODH are broad-spectrum antivirals against RNA viruses including newly-emerged coronavirus SARS-CoV-2. Protein Cell 2020; 11:723-739. [PMID: 32754890 PMCID: PMC7402641 DOI: 10.1007/s13238-020-00768-w] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 07/05/2020] [Indexed: 12/28/2022] Open
Abstract
Emerging and re-emerging RNA viruses occasionally cause epidemics and pandemics worldwide, such as the on-going outbreak of the novel coronavirus SARS-CoV-2. Herein, we identified two potent inhibitors of human DHODH, S312 and S416, with favorable drug-likeness and pharmacokinetic profiles, which all showed broad-spectrum antiviral effects against various RNA viruses, including influenza A virus, Zika virus, Ebola virus, and particularly against SARS-CoV-2. Notably, S416 is reported to be the most potent inhibitor so far with an EC50 of 17 nmol/L and an SI value of 10,505.88 in infected cells. Our results are the first to validate that DHODH is an attractive host target through high antiviral efficacy in vivo and low virus replication in DHODH knock-out cells. This work demonstrates that both S312/S416 and old drugs (Leflunomide/Teriflunomide) with dual actions of antiviral and immuno-regulation may have clinical potentials to cure SARS-CoV-2 or other RNA viruses circulating worldwide, no matter such viruses are mutated or not.
Collapse
Affiliation(s)
- Rui Xiong
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Leike Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Shiliang Li
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yuan Sun
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Minyi Ding
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yong Wang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Yongliang Zhao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Yan Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Weijuan Shang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xiaming Jiang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jiwei Shan
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Zihao Shen
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yi Tong
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Liuxin Xu
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yu Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Yingle Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Gang Zou
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Dimitri Lavillete
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zhenjiang Zhao
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Rui Wang
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Lili Zhu
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Gengfu Xiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Ke Lan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Honglin Li
- Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| | - Ke Xu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
19
|
Haddow AD, Perez-Sautu U, Wiley MR, Miller LJ, Kimmel AE, Principe LM, Wollen-Roberts SE, Shamblin JD, Valdez SM, Cazares LH, Pratt WD, Rossi FD, Lugo-Roman L, Bavari S, Palacios GF, Nalca A, Nasar F, Pitt MLM. Modeling mosquito-borne and sexual transmission of Zika virus in an enzootic host, the African green monkey. PLoS Negl Trop Dis 2020; 14:e0008107. [PMID: 32569276 PMCID: PMC7343349 DOI: 10.1371/journal.pntd.0008107] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 07/08/2020] [Accepted: 02/01/2020] [Indexed: 01/08/2023] Open
Abstract
Mosquito-borne and sexual transmission of Zika virus (ZIKV), a TORCH pathogen, recently initiated a series of large epidemics throughout the Tropics. Animal models are necessary to determine transmission risk and study pathogenesis, as well screen antivirals and vaccine candidates. In this study, we modeled mosquito and sexual transmission of ZIKV in the African green monkey (AGM). Following subcutaneous, intravaginal or intrarectal inoculation of AGMs with ZIKV, we determined the transmission potential and infection dynamics of the virus. AGMs inoculated by all three transmission routes exhibited viremia and viral shedding followed by strong virus neutralizing antibody responses, in the absence of clinical illness. All four of the subcutaneously inoculated AGMs became infected (mean peak viremia: 2.9 log10 PFU/mL, mean duration: 4.3 days) and vRNA was detected in their oral swabs, with infectious virus being detected in a subset of these specimens. Although all four of the intravaginally inoculated AGMs developed virus neutralizing antibody responses, only three had detectable viremia (mean peak viremia: 4.0 log10 PFU/mL, mean duration: 3.0 days). These three AGMs also had vRNA and infectious virus detected in both oral and vaginal swabs. Two of the four intrarectally inoculated AGMs became infected (mean peak viremia: 3.8 log10 PFU/mL, mean duration: 3.5 days). vRNA was detected in oral swabs collected from both of these infected AGMs, and infectious virus was detected in an oral swab from one of these AGMs. Notably, vRNA and infectious virus were detected in vaginal swabs collected from the infected female AGM (peak viral load: 7.5 log10 copies/mL, peak titer: 3.8 log10 PFU/mL, range of detection: 5-21 days post infection). Abnormal clinical chemistry and hematology results were detected and acute lymphadenopathy was observed in some AGMs. Infection dynamics in all three AGM ZIKV models are similar to those reported in the majority of human ZIKV infections. Our results indicate that the AGM can be used as a surrogate to model mosquito or sexual ZIKV transmission and infection. Furthermore, our results suggest that AGMs are likely involved in the enzootic maintenance and amplification cycle of ZIKV.
Collapse
Affiliation(s)
- Andrew D. Haddow
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Unai Perez-Sautu
- Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Michael R. Wiley
- Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Lynn J. Miller
- Veterinary Medicine Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Adrienne E. Kimmel
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Lucia M. Principe
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Suzanne E. Wollen-Roberts
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Joshua D. Shamblin
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Stephanie M. Valdez
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Lisa H. Cazares
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - William D. Pratt
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Franco D. Rossi
- Aerobiology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Luis Lugo-Roman
- Veterinary Medicine Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Sina Bavari
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Gustavo F. Palacios
- Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Aysegul Nalca
- Aerobiology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Farooq Nasar
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - M. Louise M. Pitt
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| |
Collapse
|
20
|
Maternal Zika Virus (ZIKV) Infection following Vaginal Inoculation with ZIKV-Infected Semen in Timed-Pregnant Olive Baboons. J Virol 2020; 94:JVI.00058-20. [PMID: 32188737 PMCID: PMC7269433 DOI: 10.1128/jvi.00058-20] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/14/2020] [Indexed: 12/15/2022] Open
Abstract
Zika virus (ZIKV) infection is now firmly linked to congenital Zika syndrome (CZS), including fetal microcephaly. While Aedes species of mosquito are the primary vector for ZIKV, sexual transmission of ZIKV is a significant route of infection. ZIKV has been documented in human, mouse, and nonhuman primate (NHP) semen. It is critical to establish NHP models of the vertical transfer of ZIKV that recapitulate human pathogenesis. We hypothesized that vaginal deposition of ZIKV-infected baboon semen would lead to maternal infection and vertical transfer in the olive baboon (Papio anubis). Epidemiological studies suggest an increased rate of CZS in the Americas compared to the original link to CZS in French Polynesia; therefore, we also compared the French Polynesian (FP) ZIKV isolate to the Puerto Rican (PR) isolate. Timed-pregnant baboons (n = 6) were inoculated via vaginal deposition of baboon semen containing 106 focus-forming units (FFU) of ZIKV (n = 3 for FP isolate H/PF/2013; n = 3 for PR isolate PRVABC59) at midgestation (86 to 95 days of gestation [dG]; term, 183 dG) on day 0 (all dams) and then at 7-day intervals through 3 weeks. Maternal blood, saliva, and cervicovaginal wash (CVW) samples were obtained. Animals were euthanized at 28 days (n = 5) or 39 days (n = 1) after the initial inoculation, and maternal/fetal tissues were collected. Viremia was achieved in 3/3 FP ZIKV-infected dams and 2/3 PR ZIKV-infected dams. ZIKV RNA was detected in CVW samples of 5/6 dams. ZIKV RNA was detected in lymph nodes but not the ovaries, uterus, cervix, or vagina in FP isolate-infected dams. ZIKV RNA was detected in lymph nodes (3/3), uterus (2/3), and vagina (2/3) in PR isolate-infected dams. Placenta, amniotic fluid, and fetal tissues were ZIKV RNA negative in the FP isolate-infected dams, whereas 2/3 PR isolate-infected dam placentas were ZIKV RNA positive. We conclude that ZIKV-infected semen is a means of ZIKV transmission during pregnancy in primates. The PR isolate appeared more capable of widespread dissemination to tissues, including reproductive tissues and placenta, than the FP isolate.IMPORTANCE Zika virus remains a worldwide health threat, with outbreaks still occurring in the Americas. While mosquitos are the primary vector for the spread of the virus, sexual transmission of Zika virus is also a significant means of infection, especially in terms of passage from an infected to an uninfected partner. While sexual transmission has been documented in humans, and male-to-female transmission has been reported in mice, ours is the first study in nonhuman primates to demonstrate infection via vaginal deposition of Zika virus-infected semen. The latter is important since a recent publication indicated that human semen inhibited, in a laboratory setting, Zika virus infection of reproductive tissues. We also found that compared to the French Polynesian isolate, the Puerto Rican Zika virus isolate led to greater spread throughout the body, particularly in reproductive tissues. The American isolates of Zika virus appear to have acquired mutations that increase their efficacy.
Collapse
|
21
|
Wang T, Li P, Zhang Y, Liu Y, Tan Z, Sun J, Ke X, Miao Y, Luo D, Hu Q, Xu F, Wang H, Zheng Z. In vivo imaging of Zika virus reveals dynamics of viral invasion in immune-sheltered tissues and vertical propagation during pregnancy. Am J Cancer Res 2020; 10:6430-6447. [PMID: 32483462 PMCID: PMC7255039 DOI: 10.7150/thno.43177] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/01/2020] [Indexed: 12/11/2022] Open
Abstract
Rationale: Zika virus (ZIKV) is a pathogenic virus known to cause a wide range of congenital abnormalities, including microcephaly, Guillain-Barre syndrome, meningoencephalitis, and other neurological complications, in humans. This study investigated the noninvasive detection of ZIKV infection in vivo, which is necessary for elucidating the virus's mechanisms of viral replication and pathogenesis, as well as to accelerate the development of anti-ZIKV therapeutic strategies. Methods: In this study, a recombinant ZIKV harbouring Nluc gene (ZIKV-Nluc) was designed, recovered, and purified. The levels of bioluminescence were directly correlated with viral loads in vitro and in vivo. The dynamics of ZIKV infection in A129 (interferon (IFN)-α/β receptor deficient), AG6 (IFN-α/β and IFN-γ receptor deficient), and C57BL/6 mice were characterized. Pregnant dams were infected with ZIKV-Nluc at E10 via intra footpad injection. Then, the pooled immune sera (anti-ZIKV neutralizing antibodies) #22-1 in ZIKV-Nluc virus-infected mice were visualized. Results: ZIKV-Nluc showed a high genetic stability and replicated well in cells with similar properties to the wild-type ZIKV (ZIKVwt). Striking bioluminescence signals were consistently observed in animal organs, including spleen, intestine, testis, uterus/ovary, and kidney. The ileocecal junction was found to be the crucial visceral target. Infection of pregnant dams with ZIKV-Nluc showed that ZIKV was capable of crossing the maternal-fetal barrier to infect the fetuses via vertical transmission. Furthermore, it was visualized that treatment with the pooled immune sera was found to greatly restrict the spread of the ZIKV-Nluc virus in mice. Conclusions: This study is the first to report the real-time noninvasive tracking of the progression of ZIKV invading immune-sheltered tissues and propagating vertically during pregnancy. The results demonstrate that ZIKV-Nluc represents a powerful tool for the study of the replication, dissemination, pathogenesis, and treatment of ZIKV in vitro and in vivo.
Collapse
|
22
|
Schwarz ER, Oliveira LJ, Bonfante F, Pu R, Pozor MA, Maclachlan NJ, Beachboard S, Barr KL, Long MT. Experimental Infection of Mid-Gestation Pregnant Female and Intact Male Sheep with Zika Virus. Viruses 2020; 12:v12030291. [PMID: 32156037 PMCID: PMC7150993 DOI: 10.3390/v12030291] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/24/2020] [Accepted: 03/06/2020] [Indexed: 12/11/2022] Open
Abstract
Zika virus (ZIKV) is an arbovirus that causes birth defects, persistent male infection, and sexual transmission in humans. The purpose of this study was to continue the development of an ovine ZIKV infection model; thus, two experiments were undertaken. In the first experiment, we built on previous pregnant sheep experiments by developing a mid-gestation model of ZIKV infection. Four pregnant sheep were challenged with ZIKV at 57–64 days gestation; two animals served as controls. After 13–15 days (corresponding with 70–79 days of gestation), one control and two infected animals were euthanized; the remaining animals were euthanized at 20–22 days post-infection (corresponding with 77–86 days of gestation). In the second experiment, six sexually mature, intact, male sheep were challenged with ZIKV and two animals served as controls. Infected animals were serially euthanized on days 2–6 and day 9 post-infection with the goal of isolating ZIKV from the male reproductive tract. In the mid-gestation study, virus was detected in maternal placenta and spleen, and in fetal organs, including the brains, spleens/liver, and umbilicus of infected fetuses. Fetuses from infected animals had visibly misshapen heads and morphometrics revealed significantly smaller head sizes in infected fetuses when compared to controls. Placental pathology was evident in infected dams. In the male experiment, ZIKV was detected in the spleen, liver, testes/epididymides, and accessory sex glands of infected animals. Results from both experiments indicate that mid-gestation ewes can be infected with ZIKV with subsequent disruption of fetal development and that intact male sheep are susceptible to ZIKV infection and viral dissemination and replication occurs in highly vascular tissues (including those of the male reproductive tract).
Collapse
Affiliation(s)
- Erika R. Schwarz
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA; (E.R.S.); (R.P.); (S.B.)
| | - Lilian J. Oliveira
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA;
| | - Francesco Bonfante
- Laboratory of Experimental Animal Models, Division of Comparative Biomedical Sciences, Instituto Zooprofilattico Sperimentale delle Venezie, 35020 Legnaro, Italy;
| | - Ruiyu Pu
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA; (E.R.S.); (R.P.); (S.B.)
| | - Malgorzata A. Pozor
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA;
| | - N. James Maclachlan
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA;
| | - Sarah Beachboard
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA; (E.R.S.); (R.P.); (S.B.)
| | - Kelli L. Barr
- Department of Biology, College of Arts and Sciences, Baylor University, Waco, TX 76798, USA;
| | - Maureen T. Long
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA; (E.R.S.); (R.P.); (S.B.)
- Correspondence:
| |
Collapse
|
23
|
Morelli F, Souza RP, Cruz TED, Damke GMZF, Damke E, Suehiro TT, Silva VRSD, Consolaro MEL. Zika virus infection in the genital tract of non-pregnant females: a systematic review. Rev Inst Med Trop Sao Paulo 2020; 62:e16. [PMID: 32130356 PMCID: PMC7051180 DOI: 10.1590/s1678-9946202062016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 02/03/2020] [Indexed: 12/12/2022] Open
Abstract
This review provides a general overview on the positivity and persistence of Zika virus (ZIKV) in female genital tract (FGT) of non-pregnant women and animals, as well as in cell cultures, and its influence on FGT health. We performed a systematic review based on the PRISMA statement to identify studies focused on "Zika virus" and "non-pregnant female" in PubMed, Embase, Scopus Scholar and Web of Knowledge databases of full-text papers and abstracts published in English, with no restrictions regarding the initial date of publication, up to August 2019. Our search terms yielded 625 records, that were 108 after removal of duplicates, leaving 517 items for title and abstract reviews. Of these, 475 did not meet the inclusion criteria, leaving 42 records for full-text review and resulting in the exclusion of 6 additional records. The remaining 36 met our inclusion criteria. Variations were observed regarding the presence and persistence of ZIKV in lower and upper genital samples. However, the FGT was the place in which ZIKV RNA has been detected, sometimes for relatively long periods, even after the clearance from blood and urine. In addition to the vagina and cervix, the endometrium, uterus and ovary (oocytes and follicles) could also be involved in persistent ZIKV infections. Further prospective studies are needed to assess the effect of ZIKV on FGT health.
Collapse
Affiliation(s)
- Fabrício Morelli
- Universidade Estadual de Maringá, Departamento de Análises Clínicas e Biomedicina, Programa de Pós-Graduação em Biociências e Fisiopatologia, Maringá, Paraná, Brazil
| | - Raquel Pantarotto Souza
- Universidade Estadual de Maringá, Departamento de Análises Clínicas e Biomedicina, Programa de Pós-Graduação em Biociências e Fisiopatologia, Maringá, Paraná, Brazil
| | - Taís Elisângela da Cruz
- Universidade Estadual de Maringá, Departamento de Análises Clínicas e Biomedicina, Programa de Pós-Graduação em Biociências e Fisiopatologia, Maringá, Paraná, Brazil
| | - Gabrielle Marconi Zago Ferreira Damke
- Universidade Estadual de Maringá, Departamento de Análises Clínicas e Biomedicina, Programa de Pós-Graduação em Biociências e Fisiopatologia, Maringá, Paraná, Brazil
| | - Edilson Damke
- Universidade Estadual de Maringá, Departamento de Análises Clínicas e Biomedicina, Programa de Pós-Graduação em Biociências e Fisiopatologia, Maringá, Paraná, Brazil
| | - Tamy Tuani Suehiro
- Universidade Estadual de Maringá, Departamento de Análises Clínicas e Biomedicina, Programa de Pós-Graduação em Biociências e Fisiopatologia, Maringá, Paraná, Brazil
| | - Vânia Ramos Sela da Silva
- Universidade Estadual de Maringá, Departamento de Análises Clínicas e Biomedicina, Programa de Pós-Graduação em Biociências e Fisiopatologia, Maringá, Paraná, Brazil
| | - Marcia Edilaine Lopes Consolaro
- Universidade Estadual de Maringá, Departamento de Análises Clínicas e Biomedicina, Programa de Pós-Graduação em Biociências e Fisiopatologia, Maringá, Paraná, Brazil
| |
Collapse
|
24
|
Zimmerman MG, Wrammert J, Suthar MS. Cross-Reactive Antibodies during Zika Virus Infection: Protection, Pathogenesis, and Placental Seeding. Cell Host Microbe 2020; 27:14-24. [PMID: 31917957 PMCID: PMC7802743 DOI: 10.1016/j.chom.2019.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Humoral immunity is an essential component of the protective immune response to flavivirus infection. Typically, primary infection generates a robust neutralizing antibody response that mediates viral control and protection. It is becoming increasingly apparent that secondary infection with a closely related flavivirus strain can result in immunological cross-reactivity; however, the consequences to infection outcome remain controversial. Since its introduction to Brazil in 2015, Zika virus (ZIKV) has caused an epidemic of fetal congenital malformations within the Americas. Because ZIKV is a mosquito-borne flavivirus with a high degree of sequence and structural homology to Dengue virus (DENV), the role of immunological cross-reactivity in ZIKV and DENV infections has become a great concern. In this review, we highlight contemporary findings that implicate a role for flavivirus antibodies in mediating protection, contributing to pathogenesis, and seeding the human placenta.
Collapse
Affiliation(s)
- Matthew G Zimmerman
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA
| | - Jens Wrammert
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA
| | - Mehul S Suthar
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA.
| |
Collapse
|
25
|
Block LN, Aliota MT, Friedrich TC, Schotzko ML, Mean KD, Wiepz GJ, Golos TG, Schmidt JK. Embryotoxic impact of Zika virus in a rhesus macaque in vitro implantation model†. Biol Reprod 2020; 102:806-816. [PMID: 31901091 DOI: 10.1093/biolre/ioz236] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/23/2019] [Accepted: 12/31/2019] [Indexed: 12/29/2022] Open
Abstract
Zika virus (ZIKV) infection is associated with adverse pregnancy outcomes in humans, and infection in the first trimester can lead to miscarriage and stillbirth. Vertical and sexual transmissions of ZIKV have been demonstrated, yet the impact of infection during the initial stages of pregnancy remains unexplored. Here we defined the impact of ZIKV on early embryonic and placental development with a rhesus macaque model. During in vitro fertilization (IVF), macaque gametes were inoculated with a physiologically relevant dose of 5.48log10 plaque-forming units (PFU) of Zika virus/H.sapiens-tc/PUR/2015/PRVABC59_v3c2. Exposure at fertilization did not alter blastocyst formation rates compared to controls. To determine the impact of ZIKV exposure at implantation, hatched blastocysts were incubated with 3.26log10, 4.26log10, or 5.26log10 PFU, or not exposed to ZIKV, followed by extended embryo culture for 10 days. ZIKV exposure negatively impacted attachment, growth, and survival in comparison to controls, with exposure to 5.26log10 PFU ZIKV resulting in embryonic degeneration by day 2. Embryonic secretion of pregnancy hormones was lower in ZIKV-exposed embryos. Increasing levels of infectious virus were detected in the culture media post-exposure, suggesting that the trophectoderm is susceptible to productive ZIKV infection. These results demonstrate that ZIKV exposure severely impacts the zona-free blastocyst, whereas exposure at the time of fertilization does not hinder blastocyst formation. Overall, early stages of pregnancy may be profoundly sensitive to infection and pregnancy loss, and the negative impact of ZIKV infection on pregnancy outcomes may be underestimated.
Collapse
Affiliation(s)
- Lindsey N Block
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA.,Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Matthew T Aliota
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, MN, USA
| | - Thomas C Friedrich
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA.,Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Michele L Schotzko
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Katherine D Mean
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Gregory J Wiepz
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Thaddeus G Golos
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA.,Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA and.,Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, USA
| | - Jenna Kropp Schmidt
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
26
|
Borucki MK, Collette NM, Coffey LL, Van Rompay KKA, Hwang MH, Thissen JB, Allen JE, Zemla AT. Multiscale analysis for patterns of Zika virus genotype emergence, spread, and consequence. PLoS One 2019; 14:e0225699. [PMID: 31809512 PMCID: PMC6897431 DOI: 10.1371/journal.pone.0225699] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 11/11/2019] [Indexed: 11/19/2022] Open
Abstract
The question of how Zika virus (ZIKV) changed from a seemingly mild virus to a human pathogen capable of microcephaly and sexual transmission remains unanswered. The unexpected emergence of ZIKV's pathogenicity and capacity for sexual transmission may be due to genetic changes, and future changes in phenotype may continue to occur as the virus expands its geographic range. Alternatively, the sheer size of the 2015-16 epidemic may have brought attention to a pre-existing virulent ZIKV phenotype in a highly susceptible population. Thus, it is important to identify patterns of genetic change that may yield a better understanding of ZIKV emergence and evolution. However, because ZIKV has an RNA genome and a polymerase incapable of proofreading, it undergoes rapid mutation which makes it difficult to identify combinations of mutations associated with viral emergence. As next generation sequencing technology has allowed whole genome consensus and variant sequence data to be generated for numerous virus samples, the task of analyzing these genomes for patterns of mutation has become more complex. However, understanding which combinations of mutations spread widely and become established in new geographic regions versus those that disappear relatively quickly is essential for defining the trajectory of an ongoing epidemic. In this study, multiscale analysis of the wealth of genomic data generated over the course of the epidemic combined with in vivo laboratory data allowed trends in mutations and outbreak trajectory to be assessed. Mutations were detected throughout the genome via deep sequencing, and many variants appeared in multiple samples and in some cases become consensus. Similarly, amino acids that were previously consensus in pre-outbreak samples were detected as low frequency variants in epidemic strains. Protein structural models indicate that most of the mutations associated with the epidemic transmission occur on the exposed surface of viral proteins. At the macroscale level, consensus data was organized into large and interactive databases to allow the spread of individual mutations and combinations of mutations to be visualized and assessed for temporal and geographical patterns. Thus, the use of multiscale modeling for identifying mutations or combinations of mutations that impact epidemic transmission and phenotypic impact can aid the formation of hypotheses which can then be tested using reverse genetics.
Collapse
Affiliation(s)
- Monica K. Borucki
- Physical Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - Nicole M. Collette
- Physical Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - Lark L. Coffey
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Koen K. A. Van Rompay
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
- California National Primate Research Center, University of California Davis, Davis, California, United States of America
| | - Mona H. Hwang
- Physical Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - James B. Thissen
- Physical Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - Jonathan E. Allen
- Computations Directorate, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - Adam T. Zemla
- Computations Directorate, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| |
Collapse
|
27
|
Caine EA, Scheaffer SM, Broughton DE, Salazar V, Govero J, Poddar S, Osula A, Halabi J, Skaznik-Wikiel ME, Diamond MS, Moley KH. Zika Virus Causes Acute Infection and Inflammation in the Ovary of Mice Without Apparent Defects in Fertility. J Infect Dis 2019; 220:1904-1914. [PMID: 31063544 PMCID: PMC6834068 DOI: 10.1093/infdis/jiz239] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/06/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Zika virus (ZIKV) has become a global concern because infection of pregnant mothers was linked to congenital birth defects. Zika virus is unique from other flaviviruses, because it is transmitted vertically and sexually in addition to by mosquito vectors. Prior studies in mice, nonhuman primates, and humans have shown that ZIKV targets the testis in males, resulting in persistent infection and oligospermia. However, its effects on the corresponding female gonads have not been evaluated. METHODS In this study, we assessed the effects of ZIKV on the ovary in nonpregnant mice. RESULTS During the acute phase, ZIKV productively infected the ovary causing accumulation of CD4+ and virus-specific CD8+ T cells. T cells protected against ZIKV infection in the ovary, as higher viral burden was measured in CD8-/- and TCRβδ-/- mice. Increased cell death and tissue inflammation in the ovary was observed during the acute phase of infection, but this normalized over time. CONCLUSIONS In contrast to that observed with males, minimal persistence and no long-term consequences of ZIKV infection on ovarian follicular reserve or fertility were demonstrated in this model. Thus, although ZIKV replicates in cells of the ovary and causes acute oophoritis, there is rapid resolution and no long-term effects on fertility, at least in mice.
Collapse
Affiliation(s)
- Elizabeth A Caine
- Departments of Medicine, Molecular Microbiology, Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Suzanne M Scheaffer
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri
| | - Darcy E Broughton
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri
| | - Vanessa Salazar
- Departments of Medicine, Molecular Microbiology, Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Jennifer Govero
- Departments of Medicine, Molecular Microbiology, Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Subhajit Poddar
- Departments of Medicine, Molecular Microbiology, Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Augustine Osula
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri
| | - Jacques Halabi
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri
| | | | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Kelle H Moley
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
28
|
High susceptibility, viral dynamics and persistence of South American Zika virus in New World monkey species. Sci Rep 2019; 9:14495. [PMID: 31601848 PMCID: PMC6787206 DOI: 10.1038/s41598-019-50918-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 09/16/2019] [Indexed: 12/18/2022] Open
Abstract
South American Zika virus (ZIKV) recently emerged as a novel human pathogen, linked with neurological disorders. However, comparative ZIKV infectivity studies in New World primates are lacking. Two members of the Callitrichidae family, common marmosets (Callithrix jacchus) and red-bellied tamarins (Saguinus labiatus), were highly susceptible to sub-cutaneous challenge with the Puerto Rico-origin ZIKVPRVABC59 strain. Both exhibited rapid, high, acute viraemia with early neuroinvasion (3 days) in peripheral and central nervous tissue. ZIKV RNA levels in blood and tissues were significantly higher in New World hosts compared to Old World species (Macaca mulatta, Macaca fascicularis). Tamarins and rhesus macaques exhibited loss of zonal occludens-1 (ZO-1) staining, indicative of a compromised blood-brain barrier 3 days post-ZIKV exposure. Early, widespread dissemination across multiple anatomical sites distant to the inoculation site preceded extensive ZIKV persistence after 100 days in New and Old World lineages, especially lymphoid, neurological and reproductive sites. Prolonged persistence in brain tissue has implications for otherwise resolved human ZIKV infection. High susceptibility of distinct New World species underscores possible establishment of ZIKV sylvatic cycles in primates indigenous to ZIKV endemic regions. Tamarins and marmosets represent viable New World models for ZIKV pathogenesis and therapeutic intervention studies, including vaccines, with contemporary strains.
Collapse
|
29
|
Prisant N, Joguet G, Herrmann-Stock C, Moriniere C, Pavili L, Lurel S, Bujan L. Upper and lower genital tract Zika virus screening in a large cohort of reproductive-age women during the Americas epidemic. Reprod Biomed Online 2019; 39:624-632. [PMID: 31375360 DOI: 10.1016/j.rbmo.2019.05.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/14/2019] [Accepted: 05/17/2019] [Indexed: 11/21/2022]
Abstract
RESEARCH QUESTION To determine whether there is a risk of localized Zika virus (ZIKV) infection in the upper genital tract, specifically the oocytes, follicular fluids and endometrium, in exposed and/or recently infected reproductive-age women. ZIKV is an Aedes mosquito-borne Flavivirus that can lead to birth defects and to developmental anomalies when it infects pregnant women. DESIGN Controlled observational clinical study following 179 female patients undergoing oocyte vitrification cycles in an academic fertility centre during the ZIKV epidemic in the French territories of the Americas. At the time, the French Ministry of Health issued a ban on medically-induced pregnancies. Oocyte vitrification cycles were the only means of preserving fertility options and ensuring Zika-free oocyte cryopreservation for currently exposed and/or recently infected patients. Samples of serum, urine, lower genital tract, endometrium, follicular fluid and immature oocytes were tested for ZIKV RNA (vRNA) by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Serological analysis for ZIKV antibodies was performed in succession for the duration of the study. The follow-up protocol was set up for more than 6 months post-exposure or post-onset. RESULTS No vRNA was detected in the various samples from exposed patients. Furthermore, no vRNA was found in the upper genital tracts of women with a recent (3 months) history of acute infection. CONCLUSION These findings represent evidence of a lack of vRNA persistence in the reproductive tract in ZIKV exposed and/or recently infected reproductive-age women and could help simplify current guidelines.
Collapse
Affiliation(s)
- Nadia Prisant
- Centre Hospitalier Universitaire de Pointe-à-Pitre, CCMR CECOS Caraïbes, Pôle Parents Enfants, Guadeloupe FWI, France; Department of Pathology, Sheba Medical Center, Tel HaShomer, Israel.
| | - Guillaume Joguet
- Centre Hospitalier Universitaire de Pointe-à-Pitre, CCMR CECOS Caraïbes, Pôle Parents Enfants, Guadeloupe FWI, France
| | - Cecile Herrmann-Stock
- Centre Hospitalier Universitaire de Pointe-à-Pitre, Laboratoire de Microbiologie Clinique et Environnementale, Guadeloupe FWI, France
| | - Catherine Moriniere
- Centre Hospitalier Universitaire de Pointe-à-Pitre, CCMR CECOS Caraïbes, Pôle Parents Enfants, Guadeloupe FWI, France
| | - Lynda Pavili
- Centre Hospitalier Universitaire de Pointe-à-Pitre, CCMR CECOS Caraïbes, Pôle Parents Enfants, Guadeloupe FWI, France
| | - Sylvia Lurel
- Centre Hospitalier Universitaire de Pointe-à-Pitre, CCMR CECOS Caraïbes, Pôle Parents Enfants, Guadeloupe FWI, France
| | - Louis Bujan
- Research Group on Human Fertility (EA3694), Toulouse III University and CECOS Hôpital Paule de Viguier, CHU Toulouse, France
| |
Collapse
|
30
|
Khan S, Lew I, Wu F, Fritts L, Fontaine KA, Tomar S, Trapecar M, Shehata HM, Ott M, Miller CJ, Sanjabi S. Low expression of RNA sensors impacts Zika virus infection in the lower female reproductive tract. Nat Commun 2019; 10:4344. [PMID: 31554802 PMCID: PMC6761111 DOI: 10.1038/s41467-019-12371-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 08/27/2019] [Indexed: 12/21/2022] Open
Abstract
Innate immune responses to Zika virus (ZIKV) are dampened in the lower female reproductive tract (LFRT) compared to other tissues, but the mechanism that underlies this vulnerability is poorly understood. Using tissues from uninfected and vaginally ZIKV-infected macaques and mice, we show that low basal expression of RNA-sensing pattern recognition receptors (PRRs), or their co-receptors, in the LFRT contributes to high viral replication in this tissue. In the LFRT, ZIKV sensing provides limited protection against viral replication, and the sensors are also minimally induced after vaginal infection. While IFNα/β receptor signaling offers minimal protection in the LFRT, it is required to prevent dissemination of ZIKV to other tissues, including the upper FRT. Our findings support a role for RNA-sensing PRRs in the dampened innate immunity against ZIKV in the LFRT compared to other tissues and underlie potential implications for systemic dissemination upon heterosexual transmission of ZIKV in women.
Collapse
MESH Headings
- Animals
- Female
- Gene Expression Regulation, Viral
- Genitalia, Female/immunology
- Genitalia, Female/metabolism
- Genitalia, Female/virology
- Humans
- Immunity, Innate/genetics
- Immunity, Innate/immunology
- Macaca mulatta
- Mice, Inbred C57BL
- Mice, Knockout
- RNA, Viral/genetics
- RNA, Viral/immunology
- Receptor, Interferon alpha-beta/genetics
- Receptor, Interferon alpha-beta/immunology
- Receptor, Interferon alpha-beta/metabolism
- Receptors, Pattern Recognition/genetics
- Receptors, Pattern Recognition/immunology
- Receptors, Pattern Recognition/metabolism
- Toll-Like Receptor 3/genetics
- Toll-Like Receptor 3/immunology
- Toll-Like Receptor 3/metabolism
- Vagina/immunology
- Vagina/metabolism
- Vagina/virology
- Virus Replication/genetics
- Virus Replication/immunology
- Zika Virus/genetics
- Zika Virus/immunology
- Zika Virus/physiology
- Zika Virus Infection/genetics
- Zika Virus Infection/immunology
- Zika Virus Infection/virology
Collapse
Affiliation(s)
- Shahzada Khan
- Virology and Immunology, Gladstone Institutes, San Francisco, CA, 94158, USA
| | - Irene Lew
- Virology and Immunology, Gladstone Institutes, San Francisco, CA, 94158, USA
| | - Frank Wu
- Virology and Immunology, Gladstone Institutes, San Francisco, CA, 94158, USA
| | - Linda Fritts
- Center for Comparative Medicine, University of California, Davis, Davis, CA, 95616, USA
- California National Primate Research Center, University of California, Davis, Davis, CA, 95616, USA
| | - Krystal A Fontaine
- Virology and Immunology, Gladstone Institutes, San Francisco, CA, 94158, USA
| | - Sakshi Tomar
- Virology and Immunology, Gladstone Institutes, San Francisco, CA, 94158, USA
| | - Martin Trapecar
- Virology and Immunology, Gladstone Institutes, San Francisco, CA, 94158, USA
| | - Hesham M Shehata
- Virology and Immunology, Gladstone Institutes, San Francisco, CA, 94158, USA
| | - Melanie Ott
- Virology and Immunology, Gladstone Institutes, San Francisco, CA, 94158, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Christopher J Miller
- Center for Comparative Medicine, University of California, Davis, Davis, CA, 95616, USA
- California National Primate Research Center, University of California, Davis, Davis, CA, 95616, USA
| | - Shomyseh Sanjabi
- Virology and Immunology, Gladstone Institutes, San Francisco, CA, 94158, USA.
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, 94143, USA.
- Genentech, South San Francisco, CA, 94080, USA.
| |
Collapse
|
31
|
Sherley M, Ong CW. Sexual transmission of Zika virus: a literature review. Sex Health 2019; 15:183-199. [PMID: 29268073 DOI: 10.1071/sh17046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 09/25/2017] [Indexed: 01/02/2023]
Abstract
Zika virus is an emerging health threat worldwide. A member of the yellow fever virus family, it is primarily spread by mosquitoes of the Aedes (Stegomyia) genus. Unusually for a mosquito-borne virus, sexual spread has also been reported; with cases of male-to-female, female-to-male and male-to-male sexual transmission all now published in the scientific literature, in both symptomatic and asymptomatic infection. Female-to-female sexual transmission has not yet been reported, but is biologically plausible. The extent of the effect of sexual transmission on the spread of Zika virus around the world is not well-characterised, but has particular relevance to travellers to and from non-endemic regions, and assisted reproduction services.
Collapse
Affiliation(s)
- Miranda Sherley
- Canberra Sexual Health Centre, Building 5, Level 1, Canberra Hospital, Garran, ACT. PO Box 11, Woden, ACT 2606, Australia
| | - Chong-Wei Ong
- Infectious Diseases Unit, Building 10, Level 4, Canberra Hospital, Garran, ACT. PO Box 11, Woden, ACT 2606, Australia
| |
Collapse
|
32
|
Schwarz ER, Pozor MA, Pu R, Barr KL, Beachboard SE, MacLachlan NJ, Prakoso D, Long MT. Experimental Infection of Pregnant Female Sheep with Zika Virus During Early Gestation. Viruses 2019; 11:E795. [PMID: 31470560 PMCID: PMC6784126 DOI: 10.3390/v11090795] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 08/22/2019] [Accepted: 08/27/2019] [Indexed: 12/18/2022] Open
Abstract
Zika virus (ZIKV) is a vertically and sexually transmissible virus resulting in severe congenital malformation. The goal of this study was to develop an ovine model of ZIKV infection. Between 28-35 days gestation (DG), four pregnant animals were infected with two doses of 6 × 106 PFU of ZIKV; four control animals received PBS. Animals were evaluated for 45 days (D) post-infection (PI) and necropsies were performed. Viral RNA was detected in infected ewe peripheral blood mononuclear cells (PBMC) during the first week PI; however, all fluids and tissues were negative upon culture. Anti-ZIKV IgM (1:400) and neutralizing antibodies were detected in all infected animals. Clinical disease, virus, or ZIKV antibodies were not detected in control ewes. After two weeks PI, fetal loss occurred in two infected animals, and at necropsy, three infected animals had placental petechiation and ecchymosis and one had hydramnion. Fetal morphometrics revealed smaller cranial circumference to crown-rump length ratios (p < 0.001) and relative brain weights (p = 0.038) in fetuses of infected animals compared with control fetuses. Immunophenotyping indicated an increase in B cells (p = 0.012) in infected sheep. Additionally, in vitro experiments using both adult and fetal cell lines demonstrated that ovine cells are highly permissive to ZIKV infection. In conclusion, ZIKV infection of pregnant sheep results in a change in fetal growth and gestational outcomes.
Collapse
Affiliation(s)
- Erika R Schwarz
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Malgorzata A Pozor
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Ruiyu Pu
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Kelli L Barr
- Department of Biology, Colleges of Arts and Sciences, Baylor University, Waco, TX 76798, USA
| | - Sarah E Beachboard
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA
| | - N James MacLachlan
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Dhani Prakoso
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Maureen T Long
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
33
|
Activation of Toll-like receptor 7/8 encoded by the X chromosome alters sperm motility and provides a novel simple technology for sexing sperm. PLoS Biol 2019; 17:e3000398. [PMID: 31408454 PMCID: PMC6691984 DOI: 10.1371/journal.pbio.3000398] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 07/08/2019] [Indexed: 12/21/2022] Open
Abstract
In most mammals, the male to female sex ratio of offspring is about 50% because half of the sperm contain either the Y chromosome or X chromosome. In mice, the Y chromosome encodes fewer than 700 genes, whereas the X chromosome encodes over 3,000 genes. Although overall gene expression is lower in sperm than in somatic cells, transcription is activated selectively in round spermatids. By regulating the expression of specific genes, we hypothesized that the X chromosome might exert functional differences in sperm that are usually masked during fertilization. In this study, we found that Toll-like receptors 7/8 (TLR7/8) coding the X chromosome were expressed by approximately 50% of the round spermatids in testis and in approximately 50% of the epididymal sperm. Especially, TLR7 was localized to the tail, and TLR8 was localized to the midpiece. Ligand activation of TLR7/8 selectively suppressed the mobility of the X chromosome–bearing sperm (X-sperm) but not the Y-sperm without altering sperm viability or acrosome formation. The difference in sperm motility allowed for the separation of Y-sperm from X-sperm. Following in vitro fertilization using the ligand-selected high-mobility sperm, 90% of the embryos were XY male. Likewise, 83% of the pups obtained following embryo transfer were XY males. Conversely, the TLR7/8-activated, slow mobility sperm produced embryos and pups that were 81% XX females. Therefore, the functional differences between Y-sperm and X-sperm motility were revealed and related to different gene expression patterns, specifically TLR7/8 on X-sperm. The Toll-like receptors TLR7 and TLR8 are encoded by the X chromosome and expressed in X-containing sperm but not Y-containing sperm. TLR7/8 ligands suppress the motility of X-containing sperm, indicating that this receptor can differentially affect sperm function on the basis of the sex chromosome they bear.
Collapse
|
34
|
Khaiboullina SF, Ribeiro FM, Uppal T, Martynova EV, Rizvanov AA, Verma SC. Zika Virus Transmission Through Blood Tissue Barriers. Front Microbiol 2019; 10:1465. [PMID: 31333605 PMCID: PMC6621930 DOI: 10.3389/fmicb.2019.01465] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 06/11/2019] [Indexed: 01/12/2023] Open
Abstract
The recent Zika virus (ZIKV) epidemic in the Americas and the Caribbean revealed a new deadly strain of the mosquito-borne virus, which has never been associated with previous outbreaks in Asia. For the first time, widespread ZIKV infection was shown to cause microcephaly and death of newborns, which was most likely due to the mutation acquired during the large outbreak recorded in French Polynesia in 2013–2014. Productive ZIKV replication and persistence has been demonstrated in placenta and fetal brains. Possible association between ZIKV and microcephaly and fetal death has been confirmed using immunocompetent mouse models in vitro and in vivo. Having crossed the placenta, ZIKV directly targets neural progenitor cells (NPCs) in developing human fetus and triggers apoptosis. The embryonic endothelial cells are exceptionally susceptible to ZIKV infection, which causes cell death and tissue necrosis. On the contrary, ZIKV infection does not affect the adult brain microvascular cell morphology and blood–brain barrier function. ZIKV is transmitted primarily by Aedes mosquito bite and is introduced into the placenta/blood through replication at the site of the entry. Also, virus can be transmitted through unprotected sex. Although, multiple possible routes of virus infection have been identified, the exact mechanism(s) utilized by ZIKV to cross the placenta still remain largely unknown. In this review, the current understanding of ZIKV infection and transmission through the placental and brain barriers is summarized.
Collapse
Affiliation(s)
- Svetlana F Khaiboullina
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, Reno, Reno, NV, United States.,Department of Exploratory Research, Scientific and Educational Center of Pharmaceutics, Kazan Federal University, Kazan, Russia
| | - Fabiola M Ribeiro
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Timsy Uppal
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, Reno, Reno, NV, United States
| | - Ekaterina V Martynova
- Department of Exploratory Research, Scientific and Educational Center of Pharmaceutics, Kazan Federal University, Kazan, Russia
| | - Albert A Rizvanov
- Department of Exploratory Research, Scientific and Educational Center of Pharmaceutics, Kazan Federal University, Kazan, Russia
| | - Subhash C Verma
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, Reno, Reno, NV, United States
| |
Collapse
|
35
|
Shim BS, Kwon YC, Ricciardi MJ, Stone M, Otsuka Y, Berri F, Kwal JM, Magnani DM, Jackson CB, Richard AS, Norris P, Busch M, Curry CL, Farzan M, Watkins D, Choe H. Zika Virus-Immune Plasmas from Symptomatic and Asymptomatic Individuals Enhance Zika Pathogenesis in Adult and Pregnant Mice. mBio 2019; 10:e00758-19. [PMID: 31266863 PMCID: PMC6606798 DOI: 10.1128/mbio.00758-19] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/30/2019] [Indexed: 01/02/2023] Open
Abstract
Preexisting immunity against dengue virus or West Nile virus was previously reported to mediate antibody-dependent enhancement (ADE) of Zika virus (ZIKV) infection in a mouse model. We show here that ZIKV-immune plasma samples from both symptomatic and asymptomatic individuals mediated ZIKV ADE of infection in vitro and in mice. In a lethal infection model with a viral inoculum 10 times higher, both ADE and protection were observed, depending on the amount of infused immune plasma. In a vertical-transmission model, ZIKV-immune plasma infused to timed pregnant mice increased fetal demise and decreased the body weight of surviving fetuses. Depletion of IgG from an immune plasma abolished ADE of infection, and the presence of purified IgG alone mediated ADE of infection. Higher viral loads and proinflammatory cytokines were detected in mice treated with ZIKV-immune plasma samples compared to those receiving control plasma. Together, these data show that passive immunization with homotypic ZIKV antibodies, depending on the concentration, could either worsen or limit a subsequent ZIKV infection.IMPORTANCE Antibody-dependent enhancement (ADE) of virus infection is common to many viruses and is problematic when plasma antibody levels decline to subneutralizing concentrations. ADE of infection is especially important among flaviviruses, many of which are the cause of global health problems. Recently, human plasma samples immune to heterologous flaviviruses were shown to promote Zika virus (ZIKV) infection. Here we showed in immunocompromised mouse models that homologous immune plasma samples protect mice from subsequent infection at high antibody concentrations but that they mediate ADE of infection and increase ZIKV pathogenesis in adult mice and fetal demise during pregnancy at low concentrations.
Collapse
Affiliation(s)
- Byoung-Shik Shim
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Young-Chan Kwon
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Michael J Ricciardi
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Mars Stone
- Viral Reference Laboratory and Repository Core, Blood Systems Research Institute, San Francisco, California, USA
| | - Yuka Otsuka
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Fatma Berri
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Jaclyn M Kwal
- Department of Obstetrics and Gynecology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Diogo M Magnani
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Cody B Jackson
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Audrey S Richard
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Philip Norris
- Viral Reference Laboratory and Repository Core, Blood Systems Research Institute, San Francisco, California, USA
- Laboratory Medicine and Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Michael Busch
- Viral Reference Laboratory and Repository Core, Blood Systems Research Institute, San Francisco, California, USA
- Laboratory Medicine and Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Christine L Curry
- Department of Obstetrics and Gynecology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Michael Farzan
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - David Watkins
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Hyeryun Choe
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| |
Collapse
|
36
|
Zika Virus Associated Pathology and Antigen Presence in the Testicle in the Absence of Sexual Transmission During Subacute to Chronic Infection in a Mouse Model. Sci Rep 2019; 9:8325. [PMID: 31171800 PMCID: PMC6554467 DOI: 10.1038/s41598-019-44582-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/09/2019] [Indexed: 11/09/2022] Open
Abstract
Zika virus (ZIKV) is an arboviral infection that has been shown to be sexually transmitted. The study outlined herein aims to determine if accessory sex glands and epididymal epithelial cells are sources of viral persistence in subacute and chronic ZIKV infection, and if infection of these organs is important in sexual transmission during long-term (chronic) infection. Male interferon type I receptor knockout (Ifnar-/-) mice were challenged with ZIKV and reproductive tissues were harvested 14 and 35 days post infection (DPI) for inoculation studies and 14, 35 and 70 DPI for histopathology. Artificial insemination fluid derived from epididymal flush and seminal plasma from the prostate and seminal vesicle was obtained from ZIKV inoculated and sham-infected males. Naïve interferon type I and II receptor knockout (AG129) female mice were pre-treated with progesterone and inoculated intravaginally with artificial insemination fluid from ZIKV-infected males. ZIKV RNA was detected in the artificial insemination fluid generated from epididymal flush or seminal plasma from ZIKV infected males at 14 and 35 DPI. ZIKV antigens were only detected in seminiferous tubules at 14 DPI. Epididymal epithelial cells did not show ZIKV antigen immunoreactivity at 14, 35 or 70 DPI. Severe fibrosing orchitis (end stage orchitis) was observed at 35 and 70 DPI. Mild inflammation and peri-tubular fibrosis were observed in the epididymis following clearance of virus. Viral RNA was not detected by PCR in whole blood samples of females from any intravaginal experimental group and only detected in 20% of subcutaneously inoculated animals (derived from 1 experimentally infected male) at 35 DPI. While ZIKV RNA and antigens can be detected in the male reproductive tract at 14 DPI and RNA can also be detected at 35 DPI, intravaginal inoculation of artificial insemination fluid from these time-points failed to result in viremia in naïve females inoculated intravaginally. These studies support the hypothesis that epididymal epithelial cells are critical to sexual transmission in immunocompromised mice. Additionally, acute but not chronic male reproductive tract infection with ZIKV results in infectious virus capable of being sexually transmitted in mice.
Collapse
|
37
|
Quispe Calla NE, Vicetti Miguel RD, Glick ME, Kwiek JJ, Gabriel JM, Cherpes TL. Exogenous oestrogen inhibits genital transmission of cell-associated HIV-1 in DMPA-treated humanized mice. J Int AIDS Soc 2019; 21. [PMID: 29334191 PMCID: PMC5810324 DOI: 10.1002/jia2.25063] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 01/02/2018] [Indexed: 12/11/2022] Open
Abstract
Introduction HIV affects more women than any other life‐threatening infectious agent, and most infections are sexually transmitted. HIV must breach the female genital tract mucosal barrier to establish systemic infection, and clinical studies indicate virus more easily evades this barrier in women using depot‐medroxyprogesterone acetate (DMPA) and other injectable progestins for contraception. Identifying a potential mechanism for this association, we learned DMPA promotes susceptibility of wild‐type mice to genital herpes simplex virus type 2 (HSV‐2) infection by reducing genital tissue expression of the cell‐cell adhesion molecule desmoglein‐1 (DSG‐1) and increasing genital mucosal permeability. Conversely, DMPA‐mediated increases in genital mucosal permeability and HSV‐2 susceptibility were eliminated in mice concomitantly administered exogenous oestrogen (E). To confirm and extend these findings, herein we used humanized mice to define effects of systemic DMPA and intravaginal (ivag) E administration on susceptibility to genital infection with cell‐associated HIV‐1. Methods Effects of DMPA or an intravaginal (ivag) E cream on engraftment of NOD‐scid‐IL‐2Rgcnull (NSG) mice with human peripheral blood mononuclear cells (hPBMCs) were defined with flow cytometry. Confocal microscopy was used to evaluate effects of DMPA, DMPA and E cream, or DMPA and the pharmacologically active component of the cream on vaginal tissue DSG‐1 expression and genital mucosal permeability to low molecular weight (LMW) molecules and hPBMCs. In other studies, hPBMC‐engrafted NSG mice (hPBMC‐NSG) received DMPA or DMPA and ivag E cream before genital inoculation with 106 HIV‐1‐infected hPBMCs. Mice were euthanized 10 days after infection, and plasma HIV‐1 load quantified by qRT‐PCR and splenocytes used to detect HIV‐1 p24 antigen via immunohistochemistry and infectious virus via TZM‐bl luciferase assay. Results Whereas hPBMC engraftment was unaffected by DMPA or E treatment, mice administered DMPA and E (cream or the pharmacologically active cream component) displayed greater vaginal tissue expression of DSG‐1 protein and decreased vaginal mucosal permeability to LMW molecules and hPBMCs versus DMPA‐treated mice. DMPA‐treated hPBMC‐NSG mice were also uniformly susceptible to genital transmission of cell‐associated HIV‐1, while no animal concomitantly administered DMPA and E cream acquired systemic HIV‐1 infection. Conclusion Exogenous E administration reduces susceptibility of DMPA‐treated humanized mice to genital HIV‐1 infection.
Collapse
Affiliation(s)
- Nirk E Quispe Calla
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Melissa E Glick
- The Ohio State University (OSU) College of Veterinary Medicine, Columbus, OH, USA
| | - Jesse J Kwiek
- Department of Microbiology, OSU College of Arts and Sciences, Columbus, OH, USA
| | | | - Thomas L Cherpes
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
38
|
Ohki CMY, Benazzato C, Russo FB, Beltrão-Braga PCB. Developing animal models of Zika virus infection for novel drug discovery. Expert Opin Drug Discov 2019; 14:577-589. [PMID: 30991850 DOI: 10.1080/17460441.2019.1597050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Just before the Brazilian outbreak, Zika virus was related to a mild infection, causing fever and skin rash. Congenital Zika Syndrome was first described in Brazil, causing microcephaly and malformations in newborns. Three years after the outbreak, the mechanisms of Zika pathogenesis are still not completely elucidated. Moreover, as of today, there is still no approved vaccine that can be administered to the susceptible population. Considering the unmet clinical need, animal models represent an unprecedented opportunity to study Zika pathophysiology and test drugs for the treatment and prevention of vertical transmission. Areas covered: The authors explore the current knowledge about Zika through animal models and advancements in drug discovery by highlighting drugs with the greatest potential to treat ZIKV infection and block vertical transmission. Expert opinion: Some drugs used to treat other infections have been repurposed to treat Zika infection, reducing the cost and time for clinical application. One promising example is Sofosbuvir, which protected mice models against Zika pathogenesis by preventing vertical transmission. Importantly, there is a lack on exploration on the long-term effects of Zika Congenital Syndrome, as well as the possible ways to treat its sequelae.
Collapse
Affiliation(s)
- Cristine Marie Yde Ohki
- a Department of Microbiology, Institute of Biomedical Sciences , University of São Paulo , São Paulo , Brazil
| | - Cecilia Benazzato
- a Department of Microbiology, Institute of Biomedical Sciences , University of São Paulo , São Paulo , Brazil.,b Department of Surgery, School of Veterinary Medicine , University of São Paulo , São Paulo , Brazil
| | - Fabiele Baldino Russo
- a Department of Microbiology, Institute of Biomedical Sciences , University of São Paulo , São Paulo , Brazil
| | | |
Collapse
|
39
|
Osuna CE, Whitney JB. Nonhuman Primate Models of Zika Virus Infection, Immunity, and Therapeutic Development. J Infect Dis 2019; 216:S928-S934. [PMID: 29267926 DOI: 10.1093/infdis/jix540] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Zika virus is a re-emerging flavivirus transmitted primarily by arthropod vectors. The recent devastating outbreak of Zika virus in Brazil was preceded by the slow global encroachment of this virus over many decades. To date, significant research efforts are underway to understand the spread and the unique pathogenesis of this virus; with the intent to rapidly develop vaccines and therapeutics. Several model systems have emerged to study Zika. This review will focus on the use of nonhuman primates to model Zika infection.
Collapse
Affiliation(s)
- Christa E Osuna
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston
| | - James B Whitney
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts
| |
Collapse
|
40
|
Gurung S, Reuter N, Preno A, Dubaut J, Nadeau H, Hyatt K, Singleton K, Martin A, Parks WT, Papin JF, Myers DA. Zika virus infection at mid-gestation results in fetal cerebral cortical injury and fetal death in the olive baboon. PLoS Pathog 2019; 15:e1007507. [PMID: 30657788 PMCID: PMC6355048 DOI: 10.1371/journal.ppat.1007507] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/31/2019] [Accepted: 12/05/2018] [Indexed: 11/21/2022] Open
Abstract
Zika virus (ZIKV) infection during pregnancy in humans is associated with an increased incidence of congenital anomalies including microcephaly as well as fetal death and miscarriage and collectively has been referred to as Congenital Zika Syndrome (CZS). Animal models for ZIKV infection in pregnancy have been developed including mice and non-human primates (NHPs). In macaques, fetal CZS outcomes from maternal ZIKV infection range from none to significant. In the present study we develop the olive baboon (Papio anubis), as a model for vertical transfer of ZIKV during pregnancy. Four mid-gestation, timed-pregnant baboons were inoculated with the French Polynesian ZIKV isolate (104 ffu). This study specifically focused on the acute phase of vertical transfer. Dams were terminated at 7 days post infection (dpi; n = 1), 14 dpi (n = 2) and 21 dpi (n = 1). All dams exhibited mild to moderate rash and conjunctivitis. Viremia peaked at 5–7 dpi with only one of three dams remaining mildly viremic at 14 dpi. An anti-ZIKV IgM response was observed by 14 dpi in all three dams studied to this stage, and two dams developed a neutralizing IgG response by either 14 dpi or 21 dpi, the latter included transfer of the IgG to the fetus (cord blood). A systemic inflammatory response (increased IL2, IL6, IL7, IL15, IL16) was observed in three of four dams. Vertical transfer of ZIKV to the placenta was observed in three pregnancies (n = 2 at 14 dpi and n = 1 at 21 dpi) and ZIKV was detected in fetal tissues in two pregnancies: one associated with fetal death at ~14 dpi, and the other in a viable fetus at 21 dpi. ZIKV RNA was detected in the fetal cerebral cortex and other tissues of both of these fetuses. In the fetus studied at 21 dpi with vertical transfer of virus to the CNS, the frontal cerebral cortex exhibited notable defects in radial glia, radial glial fibers, disorganized migration of immature neurons to the cortical layers, and signs of pathology in immature oligodendrocytes. In addition, indices of pronounced neuroinflammation were observed including astrogliosis, increased microglia and IL6 expression. Of interest, in one fetus examined at 14 dpi without detection of ZIKV RNA in brain and other fetal tissues, increased neuroinflammation (IL6 and microglia) was observed in the cortex. Although the placenta of the 14 dpi dam with fetal death showed considerable pathology, only minor pathology was noted in the other three placentas. ZIKV was detected immunohistochemically in two placentas (14 dpi) and one placenta at 21 dpi but not at 7 dpi. This is the first study to examine the early events of vertical transfer of ZIKV in a NHP infected at mid-gestation. The baboon thus represents an additional NHP as a model for ZIKV induced brain pathologies to contrast and compare to humans as well as other NHPs. Zika virus is endemic in the Americas, primarily spread through mosquitos and sexual contact. Zika virus infection during pregnancy in women is associated with a variety of fetal pathologies now referred to as Congenital Zika Syndrome (CZS), with the most severe pathology being fetal microcephaly. Developing model organisms that faithfully recreate Zika infection in humans is critical for future development of treatments and preventions. In our present study, we infected Olive baboons at mid-gestation with Zika virus and studied the acute period of viremia and transfer of Zika virus to the fetus during the first three weeks after infection to better understand the timing and mechanisms of transfer of ZIKV across the placenta, leading to CZS. We observed Zika virus transfer to fetuses resulting in fetal death in one pregnancy and in a second pregnancy, significant damage to the frontal cortex of the fetal brain at a critical period of neurodevelopment in primates. Thus, the baboon provides a promising new non-human primate model to further compare and contrast the consequences of Zika virus infection in pregnancy to humans and other non-human primates.
Collapse
Affiliation(s)
- Sunam Gurung
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Nicole Reuter
- Division of Comparative Medicine, Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Alisha Preno
- Division of Comparative Medicine, Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Jamie Dubaut
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Hugh Nadeau
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Kimberly Hyatt
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Krista Singleton
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Ashley Martin
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - W. Tony Parks
- Department of Pathology, University of Toronto, Toronto, Ontario, Canada
| | - James F. Papin
- Division of Comparative Medicine, Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Dean A. Myers
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- * E-mail:
| |
Collapse
|
41
|
Caine EA, Scheaffer SM, Arora N, Zaitsev K, Artyomov MN, Coyne CB, Moley KH, Diamond MS. Interferon lambda protects the female reproductive tract against Zika virus infection. Nat Commun 2019; 10:280. [PMID: 30655513 PMCID: PMC6336786 DOI: 10.1038/s41467-018-07993-2] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/30/2018] [Indexed: 02/08/2023] Open
Abstract
Although Zika virus (ZIKV) can be transmitted sexually and cause congenital birth defects, immune control mechanisms in the female reproductive tract (FRT) are not well characterized. Here we show that treatment of primary human vaginal and cervical epithelial cells with interferon (IFN)-α/β or IFN-λ induces host defense transcriptional signatures and inhibits ZIKV infection. We also assess the effects of IFNs on intravaginal infection of the FRT using ovariectomized mice treated with reproductive hormones. We find that mice receiving estradiol are protected against intravaginal ZIKV infection, independently of IFN-α/β or IFN-λ signaling. In contrast, mice lacking IFN-λ signaling sustain greater FRT infection when progesterone is administered. Exogenous IFN-λ treatment confers an antiviral effect when mice receive both estradiol and progesterone, but not progesterone alone. Our results identify a hormonal stage-dependent role for IFN-λ in controlling ZIKV infection in the FRT and suggest a path for minimizing sexual transmission of ZIKV in women.
Collapse
Affiliation(s)
- Elizabeth A Caine
- Departments of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Suzanne M Scheaffer
- Obstetrics and Gynecology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Nitin Arora
- Departments of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA
- The Center for Microbial Pathogenesis, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA
- Department of Pediatrics, The University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Konstantin Zaitsev
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Computer Technologies Department, ITMO University, St. Petersburg, 197101, Russia
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Carolyn B Coyne
- Departments of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA.
- The Center for Microbial Pathogenesis, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA.
| | - Kelle H Moley
- Obstetrics and Gynecology, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
| | - Michael S Diamond
- Departments of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
| |
Collapse
|
42
|
Amerson-Brown MH, Miller AL, Maxwell CA, White MM, Vincent KL, Bourne N, Pyles RB. Cultivated Human Vaginal Microbiome Communities Impact Zika and Herpes Simplex Virus Replication in ex vivo Vaginal Mucosal Cultures. Front Microbiol 2019; 9:3340. [PMID: 30692980 PMCID: PMC6340164 DOI: 10.3389/fmicb.2018.03340] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 12/24/2018] [Indexed: 01/07/2023] Open
Abstract
The human vaginal microbiome (VMB) is a complex bacterial community that interacts closely with vaginal epithelial cells (VECs) impacting the mucosal phenotype and its responses to pathogenic insults. The VMB and VEC relationship includes nutrient exchange and regulation of signaling molecules that controls numerous host functions and defends against invading pathogens. To better understand infection and replication of sexually transmitted viral pathogens in the human vaginal mucosa we used our ex vivo VEC multilayer culture system. We tested the hypothesis that selected VMB communities could be identified that alter the replication of sexually transmitted viruses consistent with reported clinical associations. Sterile VEC multilayer cultures or those colonized with VMB dominated by specific Lactobacillus spp., or VMB lacking lactobacilli, were infected with Zika virus, (ZIKV) a single stranded RNA virus, or Herpes Simplex Virus type 2 (HSV-2), a double stranded DNA virus. The virus was added to the apical surface of the cultured VEC multilayer to model transmission during vaginal intercourse. Viral replication was measured 48 h later by qPCR. The results indicated that VEC cultures colonized by VMB containing Staphylococcus spp., previously reported as inflammatory, significantly reduced the quantity of viral genomes produced by ZIKV. HSV-2 titers were decreased by nearly every VMB tested relative to the sterile control, although Lactobacillus spp.-dominated VMBs caused the greatest reduction in HSV-2 titer consistent with clinical observations. To explore the mechanism for reduced ZIKV titers, we investigated inflammation created by ZIKV infection, VMB colonization or pre-exposure to selected TLR agonists. Finally, expression levels of human beta defensins 1–3 were quantified in cultures infected by ZIKV and those colonized by VMBs that impacted ZIKV titers. Human beta defensins 1–3 produced by the VEC showed no association with ZIKV titers. The data presented expands the utility of this ex vivo model system providing controlled and reproducible methods to study the VMB impact on STIs and indicated an association between viral replication and specific bacterial species within the VMB.
Collapse
Affiliation(s)
- Megan H Amerson-Brown
- Graduate School of Biomedical Sciences, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Aaron L Miller
- Department of Pediatrics, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Carrie A Maxwell
- Department of Microbiology and Immunology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Mellodee M White
- Department of Pediatrics, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Kathleen L Vincent
- Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Nigel Bourne
- Department of Pediatrics, The University of Texas Medical Branch at Galveston, Galveston, TX, United States.,Department of Microbiology and Immunology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Richard B Pyles
- Department of Pediatrics, The University of Texas Medical Branch at Galveston, Galveston, TX, United States.,Department of Microbiology and Immunology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| |
Collapse
|
43
|
A 'Furry-Tale' of Zika Virus Infection: What Have We Learned from Animal Models? Viruses 2019; 11:v11010029. [PMID: 30621317 PMCID: PMC6356866 DOI: 10.3390/v11010029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/21/2018] [Accepted: 01/04/2019] [Indexed: 01/07/2023] Open
Abstract
The worldwide attention that the Zika virus (ZIKV) attracted, following its declaration as a Public Health Emergency of International concern by WHO in 2016, has led to a large collective effort by the international scientific community to understand its biology. Despite the mild symptoms caused by ZIKV in most infected people, the virus displays a number of worrying features, such as its ability to cause transplacental infection, fetal abnormalities and vector independent transmission through body fluids. In addition, the virus has been associated with the induction of Guillain-Barre syndrome in a number of infected individuals. With travelling, the virus has spread outside the original ZIKV endemic areas making it imperative to find ways to control it. Thus far, the large number of animal models developed to study ZIKV pathogenesis have proven to be valuable tools in understanding how the virus replicates and manifests itself in the host, its tissue tropism and the type of immune responses it induces. Still, vital questions, such as the molecular mechanisms of ZIKV persistence and the long-term consequences of ZIKV infection in the developing brain, remain unanswered. Here, we reviewed and discussed the major and most recent findings coming from animal studies and their implications for a ZIKV vaccine design.
Collapse
|
44
|
Azar SR, Rossi SL, Haller SH, Yun R, Huang JH, Plante JA, Zhou J, Olano JP, Roundy CM, Hanley KA, Weaver SC, Vasilakis N. ZIKV Demonstrates Minimal Pathologic Effects and Mosquito Infectivity in Viremic Cynomolgus Macaques. Viruses 2018; 10:v10110661. [PMID: 30469417 PMCID: PMC6267344 DOI: 10.3390/v10110661] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/13/2018] [Accepted: 11/19/2018] [Indexed: 12/15/2022] Open
Abstract
To evaluate the effects of ZIKV infection on non-human primates (NHPs), as well as to investigate whether these NHPs develop sufficient viremia to infect the major urban vector mosquito, Aedes aegypti, four cynomolgus macaques (Macaca fascicularis) were subcutaneously infected with 5.0 log10 focus-forming units (FFU) of DNA clone-derived ZIKV strain FSS13025 (Asian lineage, Cambodia, 2010). Following infection, the animals were sampled (blood, urine, tears, and saliva), underwent daily health monitoring, and were exposed to Ae. aegypti at specified time points. All four animals developed viremia, which peaked 3⁻4 days post-infection at a maximum value of 6.9 log10 genome copies/mL. No virus was detected in urine, tears, or saliva. Infection by ZIKV caused minimal overt disease: serum biochemistry and CBC values largely fell within the normal ranges, and cytokine elevations were minimal. Strikingly, the minimally colonized population of Ae. aegypti exposed to viremic animals demonstrated a maximum infection rate of 26% during peak viremia, with two of the four macaques failing to infect a single mosquito at any time point. These data indicate that cynomolgus macaques may be an effective model for ZIKV infection of humans and highlights the relative refractoriness of Ae. aegypti for ZIKV infection at the levels of viremia observed.
Collapse
Affiliation(s)
- Sasha R Azar
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Shannan L Rossi
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA.
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Sherry H Haller
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Ruimei Yun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Jing H Huang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Jessica A Plante
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Jiehua Zhou
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Juan P Olano
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Christopher M Roundy
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Kathryn A Hanley
- Department of Biology, New Mexico State University, Las Cruces, NM 88003, USA.
| | - Scott C Weaver
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA.
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Nikos Vasilakis
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA.
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
45
|
Multimodal assessments of Zika virus immune pathophysiological responses in marmosets. Sci Rep 2018; 8:17125. [PMID: 30459473 PMCID: PMC6244230 DOI: 10.1038/s41598-018-35481-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 11/06/2018] [Indexed: 11/09/2022] Open
Abstract
Animal models that recapitulate the human pathophysiology have been developed as useful research tools. Although laboratory mice are widely used, they are phylogenetically “distant” to humans. New world monkeys, such as the common marmoset (Callithrix jacchus) have steadily gained prominence. In this report, marmosets are explored as an alternate in vivo model to investigate infection and immunity of Zika virus (ZIKV). Multimodal platforms, including ultrasound and magnetic resonance imaging (MRI), flow cytometry, and multiplex microbead immunoassays were established to comprehensively decipher immune responses and pathophysiological outcomes. While ZIKV-infected marmosets had detectable ZIKV RNA load in various body fluids, animals did not develop any observable lesions in their testes and brains as shown by ultrasound and MRI. Immune-phenotyping detected differences in the numbers of B cells, CD8+ T cells and HLADR+ NK cells during the first two weeks of infection. Neutralizing ZIKV-specific antibodies were elicited to high levels and targeted epitopes in the E protein. This study presents a one-stop-shop platform to study infection and pathophysiology in marmosets. While marmoset-specific research tools are being refined, the research values of these animals present them as a good model for immune-based therapies.
Collapse
|
46
|
Clancy CS, Van Wettere AJ, Morrey JD, Julander JG. Coitus-Free Sexual Transmission of Zika Virus in a Mouse Model. Sci Rep 2018; 8:15379. [PMID: 30337585 PMCID: PMC6194026 DOI: 10.1038/s41598-018-33528-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/25/2018] [Indexed: 01/07/2023] Open
Abstract
Zika virus (ZIKV) is an arboviral infection that may be sexually transmitted. The present study aims to determine if accessory sex glands are a potential source of infectious virus and important in sexual transmission. Male interferon type I receptor knockout (Ifnar−/−) mice were challenged subcutaneously with a Puerto Rican ZIKV isolate. Reproductive tissues were harvested seven days after viral challenge and artificial insemination fluid derived from epididymis or homogenized accessory sex glands (seminal plasma) was obtained. Naïve interferon type I and II receptor knockout (AG129) females were pre-treated with progesterone, and inoculated intravaginally with either epididymal flush or seminal plasma from ZIKV-infected males. ZIKV RNA was demonstrated in the artificial insemination fluid and ZIKV antigen was detected in epididymal epithelial cells but not within seminiferous tubules at the time of artificial insemination fluid collection. Peripheral viremia, demonstrated by ZIKV RNA in whole blood samples of females from each challenge group was observed. Infectious virus was present in both epididymal fluid and seminal plasma. These studies provide evidence of passage of virus from epididymal flush and seminal plasma to naïve females via artificial insemination and provides a model for the study of sexual transmission of ZIKV.
Collapse
Affiliation(s)
- Chad S Clancy
- Utah Veterinary Diagnostic Laboratory, School of Veterinary Medicine, Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, United States of America.,Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, 84322-5600, United States of America
| | - Arnaud J Van Wettere
- Utah Veterinary Diagnostic Laboratory, School of Veterinary Medicine, Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, United States of America
| | - John D Morrey
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, 84322-5600, United States of America
| | - Justin G Julander
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, 84322-5600, United States of America.
| |
Collapse
|
47
|
Abstract
Recent Zika virus outbreaks have been associated with severe outcomes, especially during pregnancy. A great deal of effort has been put toward understanding this virus, particularly the immune mechanisms responsible for rapid viral control in the majority of infections. Identifying and understanding the key mechanisms of immune control will provide the foundation for the development of effective vaccines and antiviral therapy. Here, we outline a mathematical modeling approach for analyzing the within-host dynamics of Zika virus, and we describe how these models can be used to understand key aspects of the viral life cycle and to predict antiviral efficacy.
Collapse
Affiliation(s)
- Katharine Best
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Alan S. Perelson
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| |
Collapse
|
48
|
O'Connor MA, Tisoncik-Go J, Lewis TB, Miller CJ, Bratt D, Moats CR, Edlefsen PT, Smedley J, Klatt NR, Gale M, Fuller DH. Early cellular innate immune responses drive Zika viral persistence and tissue tropism in pigtail macaques. Nat Commun 2018; 9:3371. [PMID: 30135445 PMCID: PMC6105614 DOI: 10.1038/s41467-018-05826-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 07/24/2018] [Indexed: 12/22/2022] Open
Abstract
The immunological and virological events that contribute to the establishment of Zika virus (ZIKV) infection in humans are unclear. Here, we show that robust cellular innate immune responses arising early in the blood and tissues in response to ZIKV infection are significantly stronger in males and correlate with increased viral persistence. In particular, early peripheral blood recruitment of plasmacytoid dendritic cells and higher production of monocyte chemoattractant protein (MCP-1) correspond with greater viral persistence and tissue dissemination. We also identify non-classical monocytes as primary in vivo targets of ZIKV infection in the blood and peripheral lymph node. These results demonstrate the potential differences in ZIKV pathogenesis between males and females and a key role for early cellular innate immune responses in the blood in viral dissemination and ZIKV pathogenesis.
Collapse
Affiliation(s)
- Megan A O'Connor
- Department of Microbiology, University of Washington, Seattle, 98195, WA, USA
- Washington National Primate Research Center, Seattle, 98121, WA, USA
| | - Jennifer Tisoncik-Go
- Department of Immunology, University of Washington, Seattle, 98109, WA, USA
- Center for Innate Immunity and Immune Disease (CIIID), University of Washington, Seattle, 98109, WA, USA
| | - Thomas B Lewis
- Department of Microbiology, University of Washington, Seattle, 98195, WA, USA
- Washington National Primate Research Center, Seattle, 98121, WA, USA
| | - Charlene J Miller
- Department of Pharmaceutics, University of Washington, Seattle, 98195, WA, USA
- Department of Pediatrics, University of Miami, Miami, 33136, FL, USA
| | - Debra Bratt
- Washington National Primate Research Center, Seattle, 98121, WA, USA
| | - Cassie R Moats
- Washington National Primate Research Center, Seattle, 98121, WA, USA
- Oregon National Primate Research Center, Hillsboro, 97006, OR, USA
| | - Paul T Edlefsen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, 98109, WA, USA
| | - Jeremy Smedley
- Washington National Primate Research Center, Seattle, 98121, WA, USA
- Oregon National Primate Research Center, Hillsboro, 97006, OR, USA
| | - Nichole R Klatt
- Washington National Primate Research Center, Seattle, 98121, WA, USA
- Department of Pharmaceutics, University of Washington, Seattle, 98195, WA, USA
- Department of Pediatrics, University of Miami, Miami, 33136, FL, USA
| | - Michael Gale
- Department of Immunology, University of Washington, Seattle, 98109, WA, USA
- Center for Innate Immunity and Immune Disease (CIIID), University of Washington, Seattle, 98109, WA, USA
| | - Deborah Heydenburg Fuller
- Department of Microbiology, University of Washington, Seattle, 98195, WA, USA.
- Washington National Primate Research Center, Seattle, 98121, WA, USA.
| |
Collapse
|
49
|
Preliminary Studies on Immune Response and Viral Pathogenesis of Zika Virus in Rhesus Macaques. Pathogens 2018; 7:pathogens7030070. [PMID: 30127237 PMCID: PMC6160936 DOI: 10.3390/pathogens7030070] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 07/18/2018] [Accepted: 08/10/2018] [Indexed: 01/08/2023] Open
Abstract
Zika Virus (ZIKV) is primarily transmitted through mosquito bites. It can also be transmitted during sexual intercourse and in utero from mother to fetus. To gain preliminary insight into ZIKV pathology and immune responses on route of transmission, rhesus macaques (RMs) were inoculated with ZIKV (PRVABC59) via intravaginal (IVAG) (n = 3) or subcutaneous (sub Q) (n = 2) routes. Systemic ZIKV infection was observed in all RMs, regardless of the route of inoculation. After 9 days postinfection (dpi), ZIKV was not detected in the plasma of IVAG- and sub-Q-inoculated RMs. Importantly, RMs harbored ZIKV up to 60 dpi in various anatomical locations. Of note, ZIKV was also present in several regions of the brain, including the caudate nucleus, parietal lobe, cortex, and amygdala. These observations appear to indicate that ZIKV infection may be systemic and persistent regardless of route of inoculation. In addition, we observed changes in key immune cell populations in response to ZIKV infection. Importantly, IVAG ZIKV infection of RMs is associated with increased depletion of CD11C hi myeloid cells, reduced PD-1 expression in NK cells, and elevated frequencies of Ki67+ CD8+ central memory cells as compared to sub Q ZIKV-infected RMs. These results need to interpreted with caution due to the small number of animals utilized in this study. Future studies involving large groups of animals that have been inoculated through both routes of transmission are needed to confirm our findings.
Collapse
|
50
|
Gurung S, Preno AN, Dubaut JP, Nadeau H, Hyatt K, Reuter N, Nehete B, Wolf RF, Nehete P, Dittmer DP, Myers DA, Papin JF. Translational Model of Zika Virus Disease in Baboons. J Virol 2018; 92:e00186-18. [PMID: 29875247 PMCID: PMC6069201 DOI: 10.1128/jvi.00186-18] [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: 02/01/2018] [Accepted: 05/25/2018] [Indexed: 01/23/2023] Open
Abstract
Zika virus (ZIKV) is an emerging mosquito-borne flavivirus with devastating outcomes seen recently in the Americas due to the association of maternal ZIKV infection with fetal microcephaly and other fetal malformations not previously associated with flavivirus infections. Here, we have developed the olive baboon (Papio anubis) as a nonhuman primate (NHP) translational model for the study of ZIKV pathogenesis and associated disease outcomes to contrast and compare with humans and other major NHPs, such as macaques. Following subcutaneous inoculation of adult male and nonpregnant female baboons, viremia was detected at 3 and 4 days postinfection (dpi) with the concordant presentation of a visible rash and conjunctivitis, similar to human ZIKV infection. Furthermore, virus was detected in the mucosa and cerebrospinal fluid. A robust ZIKV-specific IgM and IgG antibody response was also observed in all the animals. These data show striking similarity between humans and the olive baboon following infection with ZIKV, suggesting our model is a suitable translational NHP model to study ZIKV pathogenesis and potential therapeutics.IMPORTANCE ZIKV was first identified in 1947 in a sentinel rhesus monkey in Uganda and subsequently spread to Southeast Asia. Until 2007, only a small number of cases were reported, and ZIKV infection was relatively minor until the South Pacific and Brazilian outbreaks, where more severe outcomes were reported. Here, we present the baboon as a nonhuman primate model for contrast and comparison with other published animal models of ZIKV, such as the mouse and macaque species. Baboons breed year round and are not currently a primary nonhuman primate species used in biomedical research, making them more readily available for studies other than human immunodeficiency virus studies, which many macaque species are designated for. This, taken together with the similarities baboons have with humans, such as immunology, reproduction, genetics, and size, makes the baboon an attractive NHP model for ZIKV studies in comparison to other nonhuman primates.
Collapse
Affiliation(s)
- Sunam Gurung
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Alisha N Preno
- Division of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Jamie P Dubaut
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Hugh Nadeau
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Kimberly Hyatt
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Nicole Reuter
- Division of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Bharti Nehete
- Department of Veterinary Sciences, The University of Texas M. D. Anderson Cancer Center, Bastrop, Texas USA
| | - Roman F Wolf
- Division of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Pramod Nehete
- Department of Veterinary Sciences, The University of Texas M. D. Anderson Cancer Center, Bastrop, Texas USA
| | - Dirk P Dittmer
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Dean A Myers
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - James F Papin
- Division of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|