101
|
Zika Virus Infection in Tupaia belangeri Causes Dermatological Manifestations and Confers Protection against Secondary Infection. J Virol 2019; 93:JVI.01982-18. [PMID: 30728253 DOI: 10.1128/jvi.01982-18] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/23/2019] [Indexed: 01/05/2023] Open
Abstract
Animal models of Zika virus (ZIKV) infection have recently been established in mice, guinea pigs, and nonhuman primates. Tree shrews (Tupaia belangeri) are an emerging experimental animal in biomedical applications, but their susceptibility to ZIKV infection has not been explored. In the present study, we show that subcutaneous inoculation of ZIKV led to rapid viremia and viral secretion in saliva, as well as to typical dermatological manifestations characterized by massive diffuse skin rash on the trunk. Global transcriptomic sequencing of peripheral blood mononuclear cells isolated from ZIKV-infected animals revealed systematic gene expression changes related to the inflammatory response and dermatological manifestations. Importantly, ZIKV infection readily triggered the production of high-titer neutralizing antibodies, thus preventing secondary homologous infection in tree shrews. However, neonatal tree shrews succumbed to ZIKV challenge upon intracerebral infection. The tree shrew model described here recapitulates the most common dermatological manifestations observed in ZIKV-infected patients and may greatly facilitate the elucidation of ZIKV pathogenesis and the development of novel vaccines and therapeutics.IMPORTANCE The reemergence of Zika virus (ZIKV) has caused a global public health crisis since 2016, and there are currently no vaccines or antiviral drugs to prevent or treat ZIKV infection. However, considerable advances have been made in understanding the biology and pathogenesis of ZIKV infection. In particular, various animal models have been successfully established to mimic ZIKV infection and its associated neurological diseases and to evaluate potential countermeasures. However, the clinical symptoms in these mouse and nonhuman primate models are different from the common clinical manifestations seen in human ZIKV patients; in particular, dermatological manifestations are rarely recapitulated in these animal models. Here, we developed a new animal model of ZIKV infection in tree shrews, a rat-sized, primate-related mammal. In vitro and in vivo characterization of ZIKV infection in tree shrews established a direct link between ZIKV infection and the immune responses and dermatological manifestations. The tree shrew model described here, as well as other available animal models, provides a valuable platform to study ZIKV pathogenesis and to evaluate vaccines and therapeutics.
Collapse
|
102
|
Rajapakse NS, Ellsworth K, Liesman RM, Ho ML, Henry N, Theel ES, Wallace A, Alvino ACI, Medeiros de Mello L, Meneses J. Unilateral Phrenic Nerve Palsy in Infants with Congenital Zika Syndrome. Emerg Infect Dis 2019; 24. [PMID: 30016248 PMCID: PMC6056128 DOI: 10.3201/eid2408.180057] [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] [Indexed: 12/19/2022] Open
Abstract
This case series of right unilateral diaphragmatic paralysis suggests peripheral nervous system involvement. Since the first identification of neonatal microcephaly cases associated with congenital Zika virus infection in Brazil in 2015, a distinctive constellation of clinical features of congenital Zika syndrome has been described. Fetal brain disruption sequence is hypothesized to underlie the devastating effects of the virus on the central nervous system. However, little is known about the effects of congenital Zika virus infection on the peripheral nervous system. We describe a series of 4 cases of right unilateral diaphragmatic paralysis in infants with congenital Zika syndrome suggesting peripheral nervous system involvement and Zika virus as a unique congenital infectious cause of this finding. All the patients described also had arthrogryposis (including talipes equinovarus) and died from complications related to progressive respiratory failure.
Collapse
|
103
|
da Silva SR, Cheng F, Huang IC, Jung JU, Gao SJ. Efficiencies and kinetics of infection in different cell types/lines by African and Asian strains of Zika virus. J Med Virol 2019; 91:179-189. [PMID: 30192399 PMCID: PMC6294704 DOI: 10.1002/jmv.25306] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 08/27/2018] [Indexed: 02/05/2023]
Abstract
After recent outbreaks, Zika virus (ZIKV) was linked to severe neurological diseases including Guillain-Barré syndrome in adults and microcephaly in newborns. The severities of pathological manifestations have been associated with different ZIKV strains. To better understand the tropism of ZIKV, we infected 10 human and four nonhuman cell lines (types) with two African (IbH30656 and MR766) and two Asian (PRVABC59 and H/FP/2013) ZIKV strains. Cell susceptibility to ZIKV infection was determined by examining viral titers, synthesis of viral proteins, and replication of positive and negative strands of viral genome. Among nonhuman cell lines, only Vero cells were efficiently infected by ZIKV. Among human cell lines, all were permissive to ZIKV infection. However, 293T and HeLa cells showed differential susceptibility towards African strains. In 293T cells, the NS1 protein was expressed at the high level by African strains but was almost not expressed by Asian strains though there was no obvious difference in viral genome replication, suggesting that the differential susceptibility might be controlled at the stage of viral protein translation. This study provides comprehensive results of the permissiveness of different cell types to both African and Asian ZIKV strains, which might help clarify their different pathogenesis.
Collapse
Affiliation(s)
- Suzane Ramos da Silva
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA
- These authors contributed equally to this work
| | - Fan Cheng
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- These authors contributed equally to this work
| | - I-Chueh Huang
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jae U. Jung
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Shou-Jiang Gao
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA
- Laboratory of Human Virology and Oncology, Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
- Corresponding author: Shou-Jiang Gao, Cancer Virology Program, UPMC Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, PA 15213; Phone: 412-623-1000; Fax: 412-623-3355;
| |
Collapse
|
104
|
Malmlov A, Bantle C, Aboellail T, Wagner K, Campbell CL, Eckley M, Chotiwan N, Gullberg RC, Perera R, Tjalkens R, Schountz T. Experimental Zika virus infection of Jamaican fruit bats (Artibeus jamaicensis) and possible entry of virus into brain via activated microglial cells. PLoS Negl Trop Dis 2019; 13:e0007071. [PMID: 30716104 PMCID: PMC6382173 DOI: 10.1371/journal.pntd.0007071] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 02/20/2019] [Accepted: 12/11/2018] [Indexed: 01/17/2023] Open
Abstract
The emergence of Zika virus (ZIKV) in the New World has led to more than 200,000 human infections. Perinatal infection can cause severe neurological complications, including fetal and neonatal microcephaly, and in adults there is an association with Guillain-Barré syndrome (GBS). ZIKV is transmitted to humans by Aedes sp. mosquitoes, yet little is known about its enzootic cycle in which transmission is thought to occur between arboreal Aedes sp. mosquitos and non-human primates. In the 1950s and '60s, several bat species were shown to be naturally and experimentally susceptible to ZIKV with acute viremia and seroconversion, and some developed neurological disease with viral antigen detected in the brain. Because of ZIKV emergence in the Americas, we sought to determine susceptibility of Jamaican fruit bats (Artibeus jamaicensis), one of the most common bats in the New World. Bats were inoculated with ZIKV PRVABC59 but did not show signs of disease. Bats held to 28 days post-inoculation (PI) had detectable antibody by ELISA and viral RNA was detected by qRT-PCR in the brain, saliva and urine in some of the bats. Immunoreactivity using polyclonal anti-ZIKV antibody was detected in testes, brain, lung and salivary glands plus scrotal skin. Tropism for mononuclear cells, including macrophages/microglia and fibroblasts, was seen in the aforementioned organs in addition to testicular Leydig cells. The virus likely localized to the brain via infection of Iba1+ macrophage/microglial cells. Jamaican fruit bats, therefore, may be a useful animal model for the study of ZIKV infection. This work also raises the possibility that bats may have a role in Zika virus ecology in endemic regions, and that ZIKV may pose a wildlife disease threat to bat populations.
Collapse
Affiliation(s)
- Ashley Malmlov
- Arthropod-Borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Collin Bantle
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, United States of America
| | - Tawfik Aboellail
- Veterinary Diagnostic Laboratories, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Kaitlyn Wagner
- Arthropod-Borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Corey L. Campbell
- Arthropod-Borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Miles Eckley
- Arthropod-Borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Nunya Chotiwan
- Arthropod-Borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Rebekah C. Gullberg
- Arthropod-Borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Rushika Perera
- Arthropod-Borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Ronald Tjalkens
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, United States of America
| | - Tony Schountz
- Arthropod-Borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| |
Collapse
|
105
|
Tai W, Voronin D, Chen J, Bao W, Kessler DA, Shaz B, Jiang S, Yazdanbakhsh K, Du L. Transfusion-Transmitted Zika Virus Infection in Pregnant Mice Leads to Broad Tissue Tropism With Severe Placental Damage and Fetal Demise. Front Microbiol 2019; 10:29. [PMID: 30728813 PMCID: PMC6351479 DOI: 10.3389/fmicb.2019.00029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 01/09/2019] [Indexed: 01/10/2023] Open
Abstract
Zika virus (ZIKV) infection during pregnancy can cause significant problems, particularly congenital Zika syndrome. Nevertheless, the potential deleterious consequences and associated mechanisms of transfusion-transmitted ZIKV infection on pregnant individuals and their fetuses and babies have not been investigated. Here we examined transmissibility of ZIKV through blood transfusion in ZIKV-susceptible pregnant A129 mice. Our data showed that transfused-transmitted ZIKV at the early infection stage led to significant viremia and broad tissue tropism in the pregnant recipient mice, which were not seen in those transfused with ZIKV-positive (ZIKV+) plasma at later infection stages. Importantly, pregnant mice transfused with early-stage, but not later stages, ZIKV+ plasma also exhibited severe placental infection with vascular damage and apoptosis, fetal infection and fetal damage, accompanied by fetal and pup death. Overall, this study suggests that transfusion-related transmission of ZIKV during initial stage of infection, which harbors high plasma viral titers, can cause serious adverse complications in the pregnant recipients and their fetuses and babies.
Collapse
Affiliation(s)
- Wanbo Tai
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, United States
| | - Denis Voronin
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, United States
| | - Jiawei Chen
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, United States
| | - Weili Bao
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, United States
| | - Debra A Kessler
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, United States
| | - Beth Shaz
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, United States
| | - Shibo Jiang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, United States.,Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai, China
| | - Karina Yazdanbakhsh
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, United States
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, United States
| |
Collapse
|
106
|
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: 52] [Impact Index Per Article: 8.7] [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
|
107
|
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
|
108
|
Contemporary Zika Virus Isolates Induce More dsRNA and Produce More Negative-Strand Intermediate in Human Astrocytoma Cells. Viruses 2018; 10:v10120728. [PMID: 30572570 PMCID: PMC6316034 DOI: 10.3390/v10120728] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 01/01/2023] Open
Abstract
The recent emergence and rapid geographic expansion of Zika virus (ZIKV) poses a significant challenge for public health. Although historically causing only mild febrile illness, recent ZIKV outbreaks have been associated with more severe neurological complications, such as Guillain-Barré syndrome and fetal microcephaly. Here we demonstrate that two contemporary (2015) ZIKV isolates from Puerto Rico and Brazil may have increased replicative fitness in human astrocytoma cells. Over a single infectious cycle, the Brazilian isolate replicates to higher titers and induces more severe cytopathic effects in human astrocytoma cells than the historical African reference strain or an early Asian lineage isolate. In addition, both contemporary isolates induce significantly more double-stranded RNA in infected astrocytoma cells, despite similar numbers of infected cells across isolates. Moreover, when we quantified positive- and negative-strand viral RNA, we found that the Asian lineage isolates displayed substantially more negative-strand replicative intermediates than the African lineage isolate in human astrocytoma cells. However, over multiple rounds of infection, the contemporary ZIKV isolates appear to be impaired in cell spread, infecting a lower proportion of cells at a low MOI despite replicating to similar or higher titers. Taken together, our data suggests that contemporary ZIKV isolates may have evolved mechanisms that allow them to replicate with increased efficiency in certain cell types, thereby highlighting the importance of cell-intrinsic factors in studies of viral replicative fitness.
Collapse
|
109
|
Tricot T, Helsen N, Kaptein SJF, Neyts J, Verfaillie CM. Human stem cell-derived hepatocyte-like cells support Zika virus replication and provide a relevant model to assess the efficacy of potential antivirals. PLoS One 2018; 13:e0209097. [PMID: 30566505 PMCID: PMC6300258 DOI: 10.1371/journal.pone.0209097] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/29/2018] [Indexed: 01/30/2023] Open
Abstract
Zika virus (ZIKV) infection during pregnancy has been extensively linked to microcephaly in newborns. High levels of ZIKV RNA were, however, also detected in mice and non-human primates in organs other than the brain, such as the liver. As ZIKV is a flavivirus closely related to the dengue and yellow fever virus, which are known to cause hepatitis, we here examined whether human hepatocytes are susceptible to ZIKV infection. We demonstrated that both human pluripotent stem cell (hPSC)-derived hepatocyte-like cells (HLCs) and the Huh7 hepatoma cell line support the complete ZIKV replication cycle. Of three antiviral molecules that inhibit ZIKV infection in Vero cells, only 7-deaza-2'-C-methyladenosine (7DMA) inhibited ZIKV replication in hPSC-HLCs, while all drugs inhibited ZIKV infection in Huh7 cells. ZIKV-infected hPSC-HLCs but not Huh7 cells mounted an innate immune and NFκβ response, which may explain the more extensive cytopathic effect observed in Huh7 cells. In conclusion, ZIKV productively infects human hepatocytes in vitro. However, significant differences in the innate immune response against ZIKV and antiviral drug sensitivity were observed when comparing hPSC-HLCs and hepatoma cells, highlighting the need to assess ZIKV infection as well as antiviral activity not only in hepatoma cells, but also in more physiologically relevant systems.
Collapse
Affiliation(s)
- Tine Tricot
- Stem Cell Institute, University of Leuven (KU Leuven), Leuven, Belgium
| | - Nicky Helsen
- Stem Cell Institute, University of Leuven (KU Leuven), Leuven, Belgium
| | - Suzanne J F Kaptein
- KU Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Johan Neyts
- KU Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | | |
Collapse
|
110
|
Pena LJ, Miranda Guarines K, Duarte Silva AJ, Sales Leal LR, Mendes Félix D, Silva A, de Oliveira SA, Junqueira Ayres CF, Júnior AS, de Freitas AC. In vitro and in vivo models for studying Zika virus biology. J Gen Virol 2018; 99:1529-1550. [DOI: 10.1099/jgv.0.001153] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Lindomar José Pena
- 1Department of Virology, Aggeu Magalhaes Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil
| | - Klarissa Miranda Guarines
- 1Department of Virology, Aggeu Magalhaes Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil
| | - Anna Jéssica Duarte Silva
- 2Department of Genetics, Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Center of Biological Sciences, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| | - Lígia Rosa Sales Leal
- 2Department of Genetics, Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Center of Biological Sciences, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| | - Daniele Mendes Félix
- 1Department of Virology, Aggeu Magalhaes Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil
| | - Adalúcia Silva
- 1Department of Virology, Aggeu Magalhaes Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil
| | - Sheilla Andrade de Oliveira
- 3Department of Immunology, Aggeu Magalhaes Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil
| | | | - Abelardo Silva Júnior
- 5Department of Veterinary Medicine, Federal University of Viçosa (UFV), Viçosa, Minas Gerais, Brazil
| | - Antonio Carlos de Freitas
- 2Department of Genetics, Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Center of Biological Sciences, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| |
Collapse
|
111
|
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
|
112
|
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
|
113
|
Heffron AS, Mohr EL, Baker D, Haj AK, Buechler CR, Bailey A, Dudley DM, Newman CM, Mohns MS, Koenig M, Breitbach ME, Rasheed M, Stewart LM, Eickhoff J, Pinapati RS, Beckman E, Li H, Patel J, Tan JC, O’Connor DH. Antibody responses to Zika virus proteins in pregnant and non-pregnant macaques. PLoS Negl Trop Dis 2018; 12:e0006903. [PMID: 30481182 PMCID: PMC6286021 DOI: 10.1371/journal.pntd.0006903] [Citation(s) in RCA: 12] [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: 08/10/2018] [Revised: 12/07/2018] [Accepted: 10/04/2018] [Indexed: 12/11/2022] Open
Abstract
The specificity of the antibody response against Zika virus (ZIKV) is not well-characterized. This is due, in part, to the antigenic similarity between ZIKV and closely related dengue virus (DENV) serotypes. Since these and other similar viruses co-circulate, are spread by the same mosquito species, and can cause similar acute clinical syndromes, it is difficult to disentangle ZIKV-specific antibody responses from responses to closely-related arboviruses in humans. Here we use high-density peptide microarrays to profile anti-ZIKV antibody reactivity in pregnant and non-pregnant macaque monkeys with known exposure histories and compare these results to reactivity following DENV infection. We also compare cross-reactive binding of ZIKV-immune sera to the full proteomes of 28 arboviruses. We independently confirm a purported ZIKV-specific IgG antibody response targeting ZIKV nonstructural protein 2B (NS2B) that was recently reported in ZIKV-infected people and we show that antibody reactivity in pregnant animals can be detected as late as 127 days post-infection (dpi). However, we also show that these responses wane over time, sometimes rapidly, and in one case the response was elicited following DENV infection in a previously ZIKV-exposed animal. These results suggest epidemiologic studies assessing seroprevalence of ZIKV immunity using linear epitope-based strategies will remain challenging to interpret due to susceptibility to false positive results. However, the method used here demonstrates the potential for rapid profiling of proteome-wide antibody responses to a myriad of neglected diseases simultaneously and may be especially useful for distinguishing antibody reactivity among closely related pathogens.
Collapse
Affiliation(s)
- Anna S. Heffron
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Emma L. Mohr
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States of America
| | - David Baker
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Amelia K. Haj
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Connor R. Buechler
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Adam Bailey
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Dawn M. Dudley
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Christina M. Newman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Mariel S. Mohns
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Michelle Koenig
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Meghan E. Breitbach
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Mustafa Rasheed
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Laurel M. Stewart
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Jens Eickhoff
- Department of Biostatistics & Medical Informatics, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Richard S. Pinapati
- Technology Innovation, Roche Sequencing Solutions, Madison, WI, United States of America
| | - Erica Beckman
- Technology Innovation, Roche Sequencing Solutions, Madison, WI, United States of America
| | - Hanying Li
- Technology Innovation, Roche Sequencing Solutions, Madison, WI, United States of America
| | - Jigar Patel
- Technology Innovation, Roche Sequencing Solutions, Madison, WI, United States of America
| | - John C. Tan
- Technology Innovation, Roche Sequencing Solutions, Madison, WI, United States of America
| | - David H. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, United States of America
| |
Collapse
|
114
|
Terzian ACB, Zini N, Sacchetto L, Rocha RF, Parra MCP, Del Sarto JL, Dias ACF, Coutinho F, Rayra J, da Silva RA, Costa VV, Fernandes NCCDA, Réssio R, Díaz-Delgado J, Guerra J, Cunha MS, Catão-Dias JL, Bittar C, Reis AFN, Santos INPD, Ferreira ACM, Cruz LEAA, Rahal P, Ullmann L, Malossi C, Araújo JPD, Widen S, de Rezende IM, Mello É, Pacca CC, Kroon EG, Trindade G, Drumond B, Chiaravalloti-Neto F, Vasilakis N, Teixeira MM, Nogueira ML. Evidence of natural Zika virus infection in neotropical non-human primates in Brazil. Sci Rep 2018; 8:16034. [PMID: 30375482 PMCID: PMC6207778 DOI: 10.1038/s41598-018-34423-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 10/17/2018] [Indexed: 01/11/2023] Open
Abstract
In Africa, Old World Primates are involved in the maintenance of sylvatic circulation of ZIKV. However, in Brazil, the hosts for the sylvatic cycle remain unknown. We hypothesized that free-living NHPs might play a role in urban/periurban ZIKV dynamics, thus we undertook an NHP ZIKV investigation in two cities in Brazil. We identified ZIKV-positive NHPs and sequences obtained were phylogenetically related to the American lineage of ZIKV. Additionally, we inoculated four C. penicillata with ZIKV and our results demonstrated that marmosets had a sustained viremia. The natural and experimental infection of NHPs with ZIKV, support the hypothesis that NHPs may be a vertebrate host in the maintainance of ZIKV transmission/circulation in urban tropical settings. Further studies are needed to understand the role they may play in maintaining the urban cycle of the ZIKV and how they may be a conduit in establishing an enzootic transmission cycle in tropical Latin America.
Collapse
Affiliation(s)
- Ana Carolina B Terzian
- São José do Rio Preto School of Medicine (FAMERP), Avenida Brigadeiro Faria Lima, 5416, CEP: 15090-000, Vila São Pedro, São José do Rio Preto, SP, Brazil
| | - Nathalia Zini
- São José do Rio Preto School of Medicine (FAMERP), Avenida Brigadeiro Faria Lima, 5416, CEP: 15090-000, Vila São Pedro, São José do Rio Preto, SP, Brazil
| | - Lívia Sacchetto
- Laboratório de Vírus - Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, CEP: 31270-901, Pampulha, Belo Horizonte, MG, Brazil
| | - Rebeca Froes Rocha
- Center for Drug Research and Development, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, CEP: 31270-901, Pampulha, Belo Horizonte, MG, Brazil
| | - Maisa Carla Pereira Parra
- São José do Rio Preto School of Medicine (FAMERP), Avenida Brigadeiro Faria Lima, 5416, CEP: 15090-000, Vila São Pedro, São José do Rio Preto, SP, Brazil
| | - Juliana Lemos Del Sarto
- Center for Drug Research and Development, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, CEP: 31270-901, Pampulha, Belo Horizonte, MG, Brazil
| | - Ana Carolina Fialho Dias
- Center for Drug Research and Development, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, CEP: 31270-901, Pampulha, Belo Horizonte, MG, Brazil
| | - Felipe Coutinho
- Center for Drug Research and Development, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, CEP: 31270-901, Pampulha, Belo Horizonte, MG, Brazil
| | - Jéssica Rayra
- Center for Drug Research and Development, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, CEP: 31270-901, Pampulha, Belo Horizonte, MG, Brazil
| | - Rafael Alves da Silva
- São José do Rio Preto School of Medicine (FAMERP), Avenida Brigadeiro Faria Lima, 5416, CEP: 15090-000, Vila São Pedro, São José do Rio Preto, SP, Brazil
| | - Vivian Vasconcelos Costa
- Center for Drug Research and Development, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, CEP: 31270-901, Pampulha, Belo Horizonte, MG, Brazil
| | | | - Rodrigo Réssio
- Instituto Adolfo Lutz (IAL), Avenida Dr. Arnaldo, 351 - 7 Andar, Sala 706, CEP: 01246-000, Pacaembú, São Paulo, SP, Brazil
| | - Josué Díaz-Delgado
- Instituto Adolfo Lutz (IAL), Avenida Dr. Arnaldo, 351 - 7 Andar, Sala 706, CEP: 01246-000, Pacaembú, São Paulo, SP, Brazil
| | - Juliana Guerra
- Instituto Adolfo Lutz (IAL), Avenida Dr. Arnaldo, 351 - 7 Andar, Sala 706, CEP: 01246-000, Pacaembú, São Paulo, SP, Brazil
| | - Mariana S Cunha
- Instituto Adolfo Lutz (IAL), Avenida Dr. Arnaldo, 351 - 7 Andar, Sala 706, CEP: 01246-000, Pacaembú, São Paulo, SP, Brazil
| | - José Luiz Catão-Dias
- Laboratory of Wildlife Comparative Pathology, Department of Pathology, School of Veterinary Medicine and Animal Sciences, University of São Paulo (LAPOCM-FMVZ-USP), Avenida Orlando Marques de Paiva, 87, CEP: 05508-270, São Paulo, SP, Brazil
| | - Cintia Bittar
- Department of Biology, Institute of Biosciences, Letters, and Exact Sciences - São Paulo State University, São José do Rio Preto - (IBILCE/UNESP), Rua Cristóvão Colombo, 2265, CEP: 15054-000, São José do Rio Preto, SP, Brazil
| | - Andréia Francesli Negri Reis
- Epidemiological Surveillance Departament of São José do Rio Preto, Avenida Romeu Strazzi, 199, CEP: 15084-010, Vila Sinibaldi, São José do Rio Preto, SP, Brazil
| | - Izalco Nuremberg Penha Dos Santos
- Epidemiological Surveillance Departament of São José do Rio Preto, Avenida Romeu Strazzi, 199, CEP: 15084-010, Vila Sinibaldi, São José do Rio Preto, SP, Brazil
| | - Andréia Cristina Marascalchi Ferreira
- Epidemiological Surveillance Departament of São José do Rio Preto, Avenida Romeu Strazzi, 199, CEP: 15084-010, Vila Sinibaldi, São José do Rio Preto, SP, Brazil
| | - Lilian Elisa Arão Antônio Cruz
- Epidemiological Surveillance Departament of São José do Rio Preto, Avenida Romeu Strazzi, 199, CEP: 15084-010, Vila Sinibaldi, São José do Rio Preto, SP, Brazil
| | - Paula Rahal
- Department of Biology, Institute of Biosciences, Letters, and Exact Sciences - São Paulo State University, São José do Rio Preto - (IBILCE/UNESP), Rua Cristóvão Colombo, 2265, CEP: 15054-000, São José do Rio Preto, SP, Brazil
| | - Leila Ullmann
- São Paulo State University (Unesp), Institute for Biotechnology, Alameda das Tecomarias, s/n, CEP: 18607-440, Chácara Capão Bonito, Botucatu, SP, Brazil
| | - Camila Malossi
- São Paulo State University (Unesp), Institute for Biotechnology, Alameda das Tecomarias, s/n, CEP: 18607-440, Chácara Capão Bonito, Botucatu, SP, Brazil
| | - João Pessoa de Araújo
- São Paulo State University (Unesp), Institute for Biotechnology, Alameda das Tecomarias, s/n, CEP: 18607-440, Chácara Capão Bonito, Botucatu, SP, Brazil
| | - Steven Widen
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-0645, USA
| | - Izabela Maurício de Rezende
- Laboratório de Vírus - Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, CEP: 31270-901, Pampulha, Belo Horizonte, MG, Brazil
| | - Érica Mello
- Centro de Controle de Zoonoses, Belo Horizonte Council, Rua Édna Quintel, 173, CEP: 31270-705, São Bernardo, Belo Horizonte, MG, Brazil
| | - Carolina Colombelli Pacca
- Faceres Medical School, Avenida Anísio Haddad, 6751, CEP: 15090-305, Jardim Francisco Fernandes, São José do Rio Preto, SP, Brazil
| | - Erna Geessien Kroon
- Laboratório de Vírus - Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, CEP: 31270-901, Pampulha, Belo Horizonte, MG, Brazil
| | - Giliane Trindade
- Laboratório de Vírus - Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, CEP: 31270-901, Pampulha, Belo Horizonte, MG, Brazil
| | - Betânia Drumond
- Laboratório de Vírus - Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, CEP: 31270-901, Pampulha, Belo Horizonte, MG, Brazil
| | - Francisco Chiaravalloti-Neto
- Department of Epidemiology, School of Public Health of the University of São Paulo, Avenida Dr. Arnaldo, 715, CEP: 01246-904, São Paulo, SP, Brazil
| | - Nikos Vasilakis
- Department of Pathology and Center of Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Institute for Human Infections and Immunity, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-0609, USA
| | - Mauro M Teixeira
- Center for Drug Research and Development, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, CEP: 31270-901, Pampulha, Belo Horizonte, MG, Brazil
| | - Maurício Lacerda Nogueira
- São José do Rio Preto School of Medicine (FAMERP), Avenida Brigadeiro Faria Lima, 5416, CEP: 15090-000, Vila São Pedro, São José do Rio Preto, SP, Brazil.
| |
Collapse
|
115
|
Bidokhti MRM, Dutta D, Madduri LSV, Woollard SM, Norgren R, Giavedoni L, Byrareddy SN. SIV/SHIV-Zika co-infection does not alter disease pathogenesis in adult non-pregnant rhesus macaque model. PLoS Negl Trop Dis 2018; 12:e0006811. [PMID: 30359380 PMCID: PMC6201872 DOI: 10.1371/journal.pntd.0006811] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 08/31/2018] [Indexed: 11/19/2022] Open
Abstract
Due to the large geographical overlap of populations exposed to Zika virus (ZIKV) and human immunodeficiency virus (HIV), understanding the disease pathogenesis of co-infection is urgently needed. This warrants the development of an animal model for HIV-ZIKV co-infection. In this study, we used adult non-pregnant macaques that were chronically infected with simian immunodeficiency virus/chimeric simian human immunodeficiency virus (SIV/SHIV) and then inoculated with ZIKV. Plasma viral loads of both SIV/SHIV and ZIKV co-infected animals revealed no significant changes as compared to animals that were infected with ZIKV alone or as compared to SIV/SHIV infected animals prior to ZIKV inoculation. ZIKV tissue clearance of co-infected animals was similar to animals that were infected with ZIKV alone. Furthermore, in co-infected macaques, there was no statistically significant difference in plasma cytokines/chemokines levels as compared to prior to ZIKV inoculation. Collectively, these findings suggest that co-infection may not alter disease pathogenesis, thus warranting larger HIV-ZIKV epidemiological studies in order to validate these findings.
Collapse
Affiliation(s)
- Mehdi R. M. Bidokhti
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Debashis Dutta
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Lepakshe S. V. Madduri
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Shawna M. Woollard
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Robert Norgren
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Luis Giavedoni
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, United States of America
| | - Siddappa N. Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States of America
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States of America
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
| |
Collapse
|
116
|
Li B, Liao HM, Liu H, Tsai S, Zhang J, Hung GC, Chin PJ, Gao Y, Lo SC. Comparative genomics, infectivity and cytopathogenicity of Zika viruses produced by acutely and persistently infected human hematopoietic cell lines. PLoS One 2018; 13:e0203331. [PMID: 30192813 PMCID: PMC6128475 DOI: 10.1371/journal.pone.0203331] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/17/2018] [Indexed: 12/17/2022] Open
Abstract
Zika virus (ZIKV), an arthropod-borne virus, has emerged as a major human pathogen. Prolonged or persistent ZIKV infection of human cells and tissues may serve as a reservoir for the virus and present serious challenges to the safety of public health. Human hematopoietic cell lines with different developmental properties revealed differences in susceptibility and outcomes to ZIKV infection. In three separate studies involving the prototypic MR 766 ZIKV strain and the human monocytic leukemia U937 cell line, ZIKV initially developed only a low-grade infection at a slow rate. After continuous culture for several months, persistently ZIKV-infected cell lines were observed with most, if not all, cells testing positive for ZIKV antigen. The infected cultures produced ZIKV RNA (v-RNA) and infectious ZIKVs persistently (“persistent ZIKVs”) with distinct infectivity and pathogenicity when tested using various kinds of host cells. When the genomes of ZIKVs from the three persistently infected cell lines were compared with the genome of the prototypic MR 766 ZIKV strain, distinct sets of mutations specific to each cell line were found. Significantly, all three “persistent ZIKVs” were capable of infecting fresh U937 cells with high efficiency at rapid rates, resulting in the development of a new set of persistently ZIKV-infected U937 cell lines. The genomes of ZIKVs from the new set of persistently ZIKV-infected U937 cell lines were further analyzed for their different mutations. The 2nd generation of persistent ZIKVs continued to possess most of the distinct sets of mutations specific to the respective 1st generation of persistent ZIKVs. We anticipate that the study will contribute to the understanding of the fundamental biology of adaptive mutations and selection during viral persistence. The persistently ZIKV-infected human cell lines that we developed will also be useful to investigate critical molecular pathways of ZIKV persistence and to study drugs or countermeasures against ZIKV infections and transmission.
Collapse
Affiliation(s)
- Bingjie Li
- Tissue Microbiology Laboratory, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Hsiao-Mei Liao
- Tissue Microbiology Laboratory, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Hebing Liu
- Tissue Microbiology Laboratory, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Shien Tsai
- Tissue Microbiology Laboratory, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Jing Zhang
- Tissue Microbiology Laboratory, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Guo-Chiuan Hung
- Tissue Microbiology Laboratory, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Pei-Ju Chin
- Tissue Microbiology Laboratory, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Yamei Gao
- Lab of Pediatric and Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Shyh-Ching Lo
- Tissue Microbiology Laboratory, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
- * E-mail:
| |
Collapse
|
117
|
Alcendor DJ. Zika virus infection and implications for kidney disease. J Mol Med (Berl) 2018; 96:1145-1151. [PMID: 30171265 PMCID: PMC6208949 DOI: 10.1007/s00109-018-1692-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 08/21/2018] [Accepted: 08/24/2018] [Indexed: 12/22/2022]
Abstract
High-level and persistent viruria observed in patients infected by Zika virus (ZIKV) has been well documented. However, renal pathology in acutely infected, immunocompetent patients remains subclinical. Moreover, the long-term impact of ZIKV infection, replication, and persistence in the renal compartment of adults and infants as well as immunosuppressed patients and solid organ transplant (SOT) recipients is unknown. Mechanisms involving host and viral factors that limit or control ZIKV pathogenesis in the renal compartment are important yet unexplored. The observation that long-term viral shedding occurs in the renal compartment in the absence of clinical disease requires further investigation. In this review, I explore Zika virus-induced renal pathology in animal models, the dynamics of virus shedding in urine, virus replication in glomerular cells, ZIKV infection in human renal transplantation, and the potential impact of long-term persistent ZIKV infection in the renal compartment.
Collapse
Affiliation(s)
- Donald J Alcendor
- Center for AIDS Health Disparities Research, Department of Microbiology, Immunology, and Physiology, School of Medicine, Meharry Medical College, 1005 Dr. D.B. Todd Jr. Blvd., Hubbard Hospital, 5th Floor, Rm. 5025, Nashville, TN, 37208, USA.
| |
Collapse
|
118
|
The emergence of Zika virus and its new clinical syndromes. Nature 2018; 560:573-581. [PMID: 30158602 DOI: 10.1038/s41586-018-0446-y] [Citation(s) in RCA: 294] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 07/19/2018] [Indexed: 11/08/2022]
Abstract
Zika virus (ZIKV) is a mosquito-transmitted flavivirus that has emerged as a global health threat because of its potential to generate explosive epidemics and ability to cause congenital disease in the context of infection during pregnancy. Whereas much is known about the biology of related flaviviruses, the unique features of ZIKV pathogenesis, including infection of the fetus, persistence in immune-privileged sites and sexual transmission, have presented new challenges. The rapid development of cell culture and animal models has facilitated a new appreciation of ZIKV biology. This knowledge has created opportunities for the development of countermeasures, including multiple ZIKV vaccine candidates, which are advancing through clinical trials. Here we describe the recent advances that have led to a new understanding of the causes and consequences of the ZIKV epidemic.
Collapse
|
119
|
Extinction of Zika Virus and Usutu Virus by Lethal Mutagenesis Reveals Different Patterns of Sensitivity to Three Mutagenic Drugs. Antimicrob Agents Chemother 2018; 62:AAC.00380-18. [PMID: 29914957 PMCID: PMC6125542 DOI: 10.1128/aac.00380-18] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 06/12/2018] [Indexed: 01/02/2023] Open
Abstract
Flaviviruses constitute an increasing source of public health concern, with growing numbers of pathogens causing disease and geographic spread to temperate climates. Despite a large body of evidence supporting mutagenesis as a conceivable antiviral strategy, there are currently no data on the sensitivity to increased mutagenesis for Zika virus (ZIKV) and Usutu virus (USUV), two emerging flaviviral threats. Flaviviruses constitute an increasing source of public health concern, with growing numbers of pathogens causing disease and geographic spread to temperate climates. Despite a large body of evidence supporting mutagenesis as a conceivable antiviral strategy, there are currently no data on the sensitivity to increased mutagenesis for Zika virus (ZIKV) and Usutu virus (USUV), two emerging flaviviral threats. In this study, we demonstrate that both viruses are sensitive to three ribonucleosides, favipiravir, ribavirin, and 5-fluorouracil, that have shown mutagenic activity against other RNA viruses while remaining unaffected by a mutagenic deoxyribonucleoside. Serial cell culture passages of ZIKV in the presence of these compounds resulted in the rapid extinction of infectivity, suggesting elevated sensitivity to mutagenesis. USUV extinction was achieved when a 10-fold dilution was applied between every passage, but not in experiments involving undiluted virus, indicating an overall lower susceptibility than ZIKV. Although the two viruses are inhibited by the same three drugs, ZIKV is relatively more susceptive to serial passage in the presence of purine analogues (favipiravir and ribavirin), while USUV replication is suppressed more efficiently by 5-fluorouracil. These differences in sensitivity typically correlate with the increases in the mutation frequencies observed in each nucleoside treatment. These results are relevant to the development of efficient therapies based on lethal mutagenesis and support the rational selection of different mutagenic nucleosides for each pathogen. We will discuss the implications of these results to the fidelity of flavivirus replication and the design of antiviral therapies based on lethal mutagenesis.
Collapse
|
120
|
Saiz JC, Oya NJD, Blázquez AB, Escribano-Romero E, Martín-Acebes MA. Host-Directed Antivirals: A Realistic Alternative to Fight Zika Virus. Viruses 2018; 10:v10090453. [PMID: 30149598 PMCID: PMC6163279 DOI: 10.3390/v10090453] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 08/17/2018] [Accepted: 08/22/2018] [Indexed: 12/21/2022] Open
Abstract
Zika virus (ZIKV), a mosquito-borne flavivirus, was an almost neglected pathogen until its introduction in the Americas in 2015, where it has been responsible for a threat to global health, causing a great social and sanitary alarm due to its increased virulence, rapid spread, and an association with severe neurological and ophthalmological complications. Currently, no specific antiviral therapy against ZIKV is available, and treatments are palliative and mainly directed toward the relief of symptoms, such as fever and rash, by administering antipyretics, anti-histamines, and fluids for dehydration. Nevertheless, lately, search for antivirals has been a major aim in ZIKV investigations. To do so, screening of libraries from different sources, testing of natural compounds, and repurposing of drugs with known antiviral activity have allowed the identification of several antiviral candidates directed to both viral (structural proteins and enzymes) and cellular elements. Here, we present an updated review of current knowledge about anti-ZIKV strategies, focusing on host-directed antivirals as a realistic alternative to combat ZIKV infection.
Collapse
Affiliation(s)
- Juan-Carlos Saiz
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain.
| | - Nereida Jiménez de Oya
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain.
| | - Ana-Belén Blázquez
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain.
| | - Estela Escribano-Romero
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain.
| | - Miguel A Martín-Acebes
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain.
| |
Collapse
|
121
|
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
|
122
|
Zika virus infection of first-trimester human placentas: utility of an explant model of replication to evaluate correlates of immune protection ex vivo. Curr Opin Virol 2018; 27:48-56. [PMID: 29172071 DOI: 10.1016/j.coviro.2017.11.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/09/2017] [Indexed: 01/08/2023]
Abstract
The emergence of congenital Zika virus (ZIKV) disease, with its devastating effects on the fetus, has prompted development of vaccines and examination of how ZIKV breaches the maternal-fetal barrier. Infection of placental and decidual tissue explants has demonstrated cell types at the uterine-placental interface susceptible to infection and suggests routes for transmission across the placenta and amniochorionic membrane. ZIKV replicates in proliferating Hofbauer cells within chorionic villi in placentas from severe congenital infection. Explants of anchoring villi recapitulate placental architecture and early-stage development and suggest infected Hofbauer cells disseminate virus to fetal blood vessels. ZIKV infection of explants represents a surrogate human model for evaluating protection against transmission by antibodies in vaccine recipients and passive immune formulations and novel therapeutics.
Collapse
|
123
|
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
|
124
|
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
|
125
|
Bowen JR, Zimmerman MG, Suthar MS. Taking the defensive: Immune control of Zika virus infection. Virus Res 2018; 254:21-26. [PMID: 28867493 PMCID: PMC5832569 DOI: 10.1016/j.virusres.2017.08.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 08/26/2017] [Accepted: 08/28/2017] [Indexed: 02/06/2023]
Abstract
ZIKV is a neurotropic mosquito-borne flavivirus that has recently emerged in the Americas and is a pathogen of significant public health concern across the world. ZIKV was first isolated in Uganda in 1947 and remained dormant in Africa and Asia for decades, with sporadic outbreaks characterized by a mild self-limiting disease in humans. The emergence of ZIKV in the Americas corresponded with enhanced disease severity and congenital Zika syndrome, a phenotype characterized by severe microcephaly, brain anomalies, ocular anomalies, congenital contractures and neurological impairments. In less than two years, a collective effort led by the scientific research community has uncovered many new facets to the once rarely discussed ZIKV. In this review, we highlight the known immune parameters that correlate with protective immunity to ZIKV infection, including pattern recognition receptors, interferons, humoral and cell-mediated responses, as well as countermeasures utilized by ZIKV to inhibit host antiviral immune responses.
Collapse
Affiliation(s)
- James R Bowen
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, 30322, USA; Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA, 30329, USA; Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Matthew G Zimmerman
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, 30322, USA; Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA, 30329, USA; Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Mehul S Suthar
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, 30322, USA; Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA, 30329, USA; Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA.
| |
Collapse
|
126
|
Rajapakse NS, Ellsworth K, Liesman RM, Ho ML, Henry N, Theel ES, Wallace A, Alvino ACI, Medeiros de Mello L, Meneses J. Unilateral Phrenic Nerve Palsy in Infants with Congenital Zika Syndrome. Emerg Infect Dis 2018. [DOI: 10.3201/eid2408180057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
127
|
Matusali G, Houzet L, Satie AP, Mahé D, Aubry F, Couderc T, Frouard J, Bourgeau S, Bensalah K, Lavoué S, Joguet G, Bujan L, Cabié A, Avelar G, Lecuit M, Le Tortorec A, Dejucq-Rainsford N. Zika virus infects human testicular tissue and germ cells. J Clin Invest 2018; 128:4697-4710. [PMID: 30063220 DOI: 10.1172/jci121735] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/24/2018] [Indexed: 12/21/2022] Open
Abstract
Zika virus (ZIKV) is a teratogenic mosquito-borne flavivirus that can be sexually transmitted from man to woman. The finding of high viral loads and prolonged viral shedding in semen suggests that ZIKV replicates within the human male genital tract, but its target organs are unknown. Using ex vivo infection of organotypic cultures, we demonstrated here that ZIKV replicates in human testicular tissue and infects a broad range of cell types, including germ cells, which we also identified as infected in semen from ZIKV-infected donors. ZIKV had no major deleterious effect on the morphology and hormonal production of the human testis explants. Infection induced a broad antiviral response but no IFN upregulation and minimal proinflammatory response in testis explants, with no cytopathic effect. Finally, we studied ZIKV infection in mouse testis and compared it to human infection. This study provides key insights into how ZIKV may persist in semen and alter semen parameters, as well as a valuable tool for testing antiviral agents.
Collapse
Affiliation(s)
- Giulia Matusali
- Université de Rennes, Inserm, École des hautes études en santé publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) - UMR_S1085, Rennes, France
| | - Laurent Houzet
- Université de Rennes, Inserm, École des hautes études en santé publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) - UMR_S1085, Rennes, France
| | - Anne-Pascale Satie
- Université de Rennes, Inserm, École des hautes études en santé publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) - UMR_S1085, Rennes, France
| | - Dominique Mahé
- Université de Rennes, Inserm, École des hautes études en santé publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) - UMR_S1085, Rennes, France
| | - Florence Aubry
- Université de Rennes, Inserm, École des hautes études en santé publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) - UMR_S1085, Rennes, France
| | - Thérèse Couderc
- Institut Pasteur, Biology of Infection Unit, Paris, France.,Inserm U1117, Paris, France
| | - Julie Frouard
- Université de Rennes, Inserm, École des hautes études en santé publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) - UMR_S1085, Rennes, France
| | - Salomé Bourgeau
- Université de Rennes, Inserm, École des hautes études en santé publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) - UMR_S1085, Rennes, France
| | - Karim Bensalah
- Service d'Urologie, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Sylvain Lavoué
- Unité de coordination hospitalière des prélèvements d'organes et de tissus, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Guillaume Joguet
- Centre Caribéen de Médecine de la Reproduction-CECOS CHU de Pointe-à-Pitre, Pointe-à-Pitre, France
| | - Louis Bujan
- Research Group on Human Fertility EA 3694, University Paul Sabatier Toulouse III - CECOS, Hôpital Paule de Viguier, CHU Toulouse, Toulouse, France
| | - André Cabié
- Inserm Centre d'Investigation Clinique 1424, Centre Hospitalier Universitaire de Martinique, and Service de maladies infectieuses, Centre Hospitalier Universitaire de Martinique, Fort de France, France
| | - Gleide Avelar
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Marc Lecuit
- Institut Pasteur, Biology of Infection Unit, Paris, France.,Inserm U1117, Paris, France.,Paris-Descartes University, Department of Infectious Diseases and Tropical Medicine, Necker-Enfants Malades University Hospital, Paris, France
| | - Anna Le Tortorec
- Université de Rennes, Inserm, École des hautes études en santé publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) - UMR_S1085, Rennes, France
| | - Nathalie Dejucq-Rainsford
- Université de Rennes, Inserm, École des hautes études en santé publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) - UMR_S1085, Rennes, France
| |
Collapse
|
128
|
Abstract
Why certain viruses cross the physical barrier of the human placenta but others do not is incompletely understood. Over the past 20 years, we have gained deeper knowledge of intrauterine infection and routes of viral transmission. This review focuses on human viruses that replicate in the placenta, infect the fetus, and cause birth defects, including rubella virus, varicella-zoster virus, parvovirus B19, human cytomegalovirus (CMV), Zika virus (ZIKV), and hepatitis E virus type 1. Detailed discussions include ( a) the architecture of the uterine-placental interface, ( b) studies of placental explants ex vivo that provide insights into the infection and spread of CMV and ZIKV to the fetal compartment and how these viruses undermine early development, and ( c) novel treatments and vaccines that limit viral replication and have the potential to reduce dissemination, vertical transmission and the occurrence of congenital disease.
Collapse
Affiliation(s)
- Lenore Pereira
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, California 94143, USA;
| |
Collapse
|
129
|
Volpi VG, Pagani I, Ghezzi S, Iannacone M, D'Antonio M, Vicenzi E. Zika Virus Replication in Dorsal Root Ganglia Explants from Interferon Receptor1 Knockout Mice Causes Myelin Degeneration. Sci Rep 2018; 8:10166. [PMID: 29976926 PMCID: PMC6033858 DOI: 10.1038/s41598-018-28257-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 06/15/2018] [Indexed: 12/29/2022] Open
Abstract
Zika virus (ZIKV) is a neurotropic agent that targets the developing fetal brain in women infected during pregnancy. In addition to the developing central nervous system, ZIKV has been recently shown to infect cells of the peripheral nervous system (PNS), highlighting its potential to cause acute peripheral neuropathies in adults, such as Guillain-Barré Syndrome (GBS). Here we show that myelinating dorsal root ganglia (DRG) explants obtained from interferon-alpha/beta receptor knock-out mice are productively infected by ZIKV. Virus replication is cytopathic in both peripheral neurons and myelinating Schwann cells leading to myelin disruption. These results confirm and extend previous observations suggesting that the PNS is indeed a potential site of ZIKV infection, replication and cytopathicity.
Collapse
Affiliation(s)
- Vera Giulia Volpi
- Myelin Biology Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Isabel Pagani
- Viral Pathogens and Biosafety Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Ghezzi
- Viral Pathogens and Biosafety Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Matteo Iannacone
- Dynamics of Immune Responses Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Maurizio D'Antonio
- Myelin Biology Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Vicenzi
- Viral Pathogens and Biosafety Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
130
|
Dudley DM, Van Rompay KK, Coffey LL, Ardeshir A, Keesler RI, Bliss-Moreau E, Grigsby PL, Steinbach RJ, Hirsch AJ, MacAllister RP, Pecoraro HL, Colgin LM, Hodge T, Streblow DN, Tardif S, Patterson JL, Tamhankar M, Seferovic M, Aagaard KM, Martín CSS, Chiu CY, Panganiban AT, Veazey RS, Wang X, Maness NJ, Gilbert MH, Bohm RP, Adams Waldorf KM, Gale M, Rajagopal L, Hotchkiss CE, Mohr EL, Capuano SV, Simmons HA, Mejia A, Friedrich TC, Golos TG, O'Connor DH. Miscarriage and stillbirth following maternal Zika virus infection in nonhuman primates. Nat Med 2018; 24:1104-1107. [PMID: 29967348 PMCID: PMC6082723 DOI: 10.1038/s41591-018-0088-5] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 05/08/2018] [Indexed: 01/12/2023]
Abstract
Zika virus (ZIKV) infection in humans has been associated with severe congenital defects (i.e. microcephaly) and pregnancy loss. Here we show that 26% of nonhuman primates infected with Asian/American ZIKV in early gestation experienced fetal demise later in pregnancy despite few clinical signs of infection. Pregnancy loss due to asymptomatic ZIKV infection may therefore be a common but under-recognized adverse outcome related to maternal ZIKV infection.
Collapse
Affiliation(s)
- Dawn M Dudley
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Koen K Van Rompay
- California National Primate Research Center, University of California-Davis, Davis, CA, USA. .,Department of Pathology, Microbiology, and Immunology, University of California-Davis, Davis, CA, USA.
| | - Lark L Coffey
- Department of Pathology, Microbiology, and Immunology, University of California-Davis, Davis, CA, USA
| | - Amir Ardeshir
- California National Primate Research Center, University of California-Davis, Davis, CA, USA
| | - Rebekah I Keesler
- California National Primate Research Center, University of California-Davis, Davis, CA, USA
| | - Eliza Bliss-Moreau
- California National Primate Research Center, University of California-Davis, Davis, CA, USA
| | - Peta L Grigsby
- Department of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Portland, OR, USA
| | - Rosemary J Steinbach
- Department of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Portland, OR, USA
| | - Alec J Hirsch
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, USA.,Division of Pathobiology and Immunology, Oregon National Primate Research Center, Portland, OR, USA
| | - Rhonda P MacAllister
- Division of Comparative Medicine, Oregon National Primate Research Center, Portland, OR, USA
| | - Heidi L Pecoraro
- Pathology Services Unit, Division of Comparative Medicine, Oregon National Primate Research Center, Portland, OR, USA
| | - Lois M Colgin
- Pathology Services Unit, Division of Comparative Medicine, Oregon National Primate Research Center, Portland, OR, USA
| | - Travis Hodge
- Division of Comparative Medicine, Oregon National Primate Research Center, Portland, OR, USA
| | - Daniel N Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, USA. .,Division of Pathobiology and Immunology, Oregon National Primate Research Center, Portland, OR, USA.
| | - Suzette Tardif
- Southwest National Primate Research Center, Southwest Foundation for Biomedical Research, San Antonio, TX, USA.
| | - Jean L Patterson
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Manasi Tamhankar
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Maxim Seferovic
- Departments of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
| | - Kjersti M Aagaard
- Departments of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics at Baylor College of Medicine, Houston, TX, USA
| | | | - Charles Y Chiu
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA.,Department of Medicine/Infectious Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Antonito T Panganiban
- Division of Microbiology, Tulane National Primate Research Center, New Orleans, LA, USA. .,Department of Microbiology and Immunology, Tulane University Health Sciences Center, New Orleans, LA, USA.
| | - Ronald S Veazey
- Division of Comparative Pathology, Tulane National Primate Research Center, New Orleans, LA, USA
| | - Xiaolei Wang
- Division of Comparative Pathology, Tulane National Primate Research Center, New Orleans, LA, USA
| | - Nicholas J Maness
- Division of Microbiology, Tulane National Primate Research Center, New Orleans, LA, USA
| | - Margaret H Gilbert
- Division of Veterinary Medicine, Tulane National Primate Research Center, New Orleans, LA, USA
| | - Rudolf P Bohm
- Division of Veterinary Medicine, Tulane National Primate Research Center, New Orleans, LA, USA
| | | | - Michael Gale
- Department of Immunology, University of Washington, Seattle, WA, USA.,Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA, USA
| | - Lakshmi Rajagopal
- Department of Pediatrics, University of Washington, Seattle, WA, USA.,Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Charlotte E Hotchkiss
- Washington National Primate Research Center, University of Washington, Seattle, WA, USA
| | - Emma L Mohr
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Saverio V Capuano
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Heather A Simmons
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Andres Mejia
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 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
| | - Thaddeus G Golos
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA.,Department of Comparative Biosciences and Obstetrics and Gynecology, 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, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
131
|
Coffey LL, Keesler RI, Pesavento PA, Woolard K, Singapuri A, Watanabe J, Cruzen C, Christe KL, Usachenko J, Yee J, Heng VA, Bliss-Moreau E, Reader JR, von Morgenland W, Gibbons AM, Jackson K, Ardeshir A, Heimsath H, Permar S, Senthamaraikannan P, Presicce P, Kallapur SG, Linnen JM, Gao K, Orr R, MacGill T, McClure M, McFarland R, Morrison JH, Van Rompay KKA. Intraamniotic Zika virus inoculation of pregnant rhesus macaques produces fetal neurologic disease. Nat Commun 2018; 9:2414. [PMID: 29925843 PMCID: PMC6010452 DOI: 10.1038/s41467-018-04777-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 04/10/2018] [Indexed: 01/05/2023] Open
Abstract
Zika virus (ZIKV) infection of pregnant women can cause fetal microcephaly and other neurologic defects. We describe the development of a non-human primate model to better understand fetal pathogenesis. To reliably induce fetal infection at defined times, four pregnant rhesus macaques are inoculated intravenously and intraamniotically with ZIKV at gestational day (GD) 41, 50, 64, or 90, corresponding to first and second trimester of gestation. The GD41-inoculated animal, experiencing fetal death 7 days later, has high virus levels in fetal and placental tissues, implicating ZIKV as cause of death. The other three fetuses are carried to near term and euthanized; while none display gross microcephaly, all show ZIKV RNA in many tissues, especially in the brain, which exhibits calcifications and reduced neural precursor cells. Given that this model consistently recapitulates neurologic defects of human congenital Zika syndrome, it is highly relevant to unravel determinants of fetal neuropathogenesis and to explore interventions.
Collapse
Affiliation(s)
- Lark L Coffey
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, 1 Shields Avenue, Davis, CA, 95616, USA.
| | - Rebekah I Keesler
- California National Primate Research Center, University of California, 1 Shields Avenue, Davis, CA, 95616, USA
| | - Patricia A Pesavento
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, 1 Shields Avenue, Davis, CA, 95616, USA
| | - Kevin Woolard
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, 1 Shields Avenue, Davis, CA, 95616, USA
| | - Anil Singapuri
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, 1 Shields Avenue, Davis, CA, 95616, USA
| | - Jennifer Watanabe
- California National Primate Research Center, University of California, 1 Shields Avenue, Davis, CA, 95616, USA
| | - Christina Cruzen
- California National Primate Research Center, University of California, 1 Shields Avenue, Davis, CA, 95616, USA
| | - Kari L Christe
- California National Primate Research Center, University of California, 1 Shields Avenue, Davis, CA, 95616, USA
| | - Jodie Usachenko
- California National Primate Research Center, University of California, 1 Shields Avenue, Davis, CA, 95616, USA
| | - JoAnn Yee
- California National Primate Research Center, University of California, 1 Shields Avenue, Davis, CA, 95616, USA
| | - Victoria A Heng
- California National Primate Research Center, University of California, 1 Shields Avenue, Davis, CA, 95616, USA.,Donders Institute, Radboud University, Montessorilaan 3, 6525 HR, Nijmegen, The Netherlands
| | - Eliza Bliss-Moreau
- California National Primate Research Center, University of California, 1 Shields Avenue, Davis, CA, 95616, USA.,Department of Psychology, University of California, 1 Shields Avenue, Davis, CA, 95616, USA
| | - J Rachel Reader
- California National Primate Research Center, University of California, 1 Shields Avenue, Davis, CA, 95616, USA
| | - Wilhelm von Morgenland
- California National Primate Research Center, University of California, 1 Shields Avenue, Davis, CA, 95616, USA
| | - Anne M Gibbons
- California National Primate Research Center, University of California, 1 Shields Avenue, Davis, CA, 95616, USA
| | - Kenneth Jackson
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, 1 Shields Avenue, Davis, CA, 95616, USA
| | - Amir Ardeshir
- California National Primate Research Center, University of California, 1 Shields Avenue, Davis, CA, 95616, USA
| | - Holly Heimsath
- Duke Human Vaccine Institute, Duke University Medical Center, 103020, 2 Genome Court MSRBII, Durham, NC, 27710, USA
| | - Sallie Permar
- Duke Human Vaccine Institute, Duke University Medical Center, 103020, 2 Genome Court MSRBII, Durham, NC, 27710, USA
| | - Paranthaman Senthamaraikannan
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Pietro Presicce
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California, 10833 Le Conte Avenue, Los Angeles, CA, 90095, USA
| | - Suhas G Kallapur
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California, 10833 Le Conte Avenue, Los Angeles, CA, 90095, USA
| | - Jeffrey M Linnen
- Grifols Diagnostic Solutions, Inc., 10808 Willow Court, San Diego, CA, 92127, USA
| | - Kui Gao
- Grifols Diagnostic Solutions, Inc., 10808 Willow Court, San Diego, CA, 92127, USA
| | - Robert Orr
- Office of Counterterrorism and Emerging Threats, Office of the Chief Scientist, Food and Drug Administration, 25 New Hampshire Avenue, Silver Spring, MD, 20903, USA
| | - Tracy MacGill
- Office of Counterterrorism and Emerging Threats, Office of the Chief Scientist, Food and Drug Administration, 25 New Hampshire Avenue, Silver Spring, MD, 20903, USA
| | - Michelle McClure
- Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20903, USA
| | - Richard McFarland
- The Advanced Regenerative Manufacturing Institute, 400 Commercial Street, Manchester, NH, 03101, USA
| | - John H Morrison
- California National Primate Research Center, University of California, 1 Shields Avenue, Davis, CA, 95616, USA
| | - Koen K A Van Rompay
- California National Primate Research Center, University of California, 1 Shields Avenue, Davis, CA, 95616, USA.
| |
Collapse
|
132
|
Abstract
Zika virus RNA is frequently detected in the semen of men after Zika virus infection. To learn more about persistence of viruses in genital fluids, we searched PubMed for relevant articles. We found evidence that 27 viruses, across a broad range of virus families, can be found in human semen.
Collapse
|
133
|
Abstract
Humans have a close phylogenetic relationship with nonhuman primates (NHPs) and share many physiological parallels, such as highly similar immune systems, with them. Importantly, NHPs can be infected with many human or related simian viruses. In many cases, viruses replicate in the same cell types as in humans, and infections are often associated with the same pathologies. In addition, many reagents that are used to study the human immune response cross-react with NHP molecules. As such, NHPs are often used as models to study viral vaccine efficacy and antiviral therapeutic safety and efficacy and to understand aspects of viral pathogenesis. With several emerging viral infections becoming epidemic, NHPs are proving to be a very beneficial benchmark for investigating human viral infections.
Collapse
Affiliation(s)
- Jacob D Estes
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD, USA
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, USA
| | - Scott W Wong
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, USA
| | - Jason M Brenchley
- Barrier Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA.
| |
Collapse
|
134
|
Johnson TP, Nath A. Neurological syndromes driven by postinfectious processes or unrecognized persistent infections. Curr Opin Neurol 2018; 31:318-324. [PMID: 29547402 PMCID: PMC11391419 DOI: 10.1097/wco.0000000000000553] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW The immune system serves a critical role in protecting the host against various pathogens. However, under circumstances, once triggered by the infectious process, it may be detrimental to the host. This may be as a result of nonspecific immune activation or due to a targeted immune response to a specific host antigen. In this opinion piece, we discuss the underlying mechanisms that lead to such an inflammatory or autoimmune syndrome affecting the nervous system. We examine these hypotheses in the context of recent emerging infections to provide mechanistic insight into the clinical manifestations and rationale for immunomodulatory therapy. RECENT FINDINGS Some pathogens endure longer than previously thought. Persistent infections may continue to drive immune responses resulting in chronic inflammation or development of autoimmune processes, resulting in damage to the nervous system. Patients with genetic susceptibilities in immune regulation may be particularly vulnerable to pathogen driven autoimmune responses. SUMMARY The presence of prolonged pathogens may result in chronic immune stimulations that drives immune-mediated neurologic complications. Understanding the burden and mechanisms of these processes is challenging but important.
Collapse
Affiliation(s)
- Tory P Johnson
- Richard T Johnson Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore
| | - Avindra Nath
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
135
|
Bosch I, de Puig H, Hiley M, Carré-Camps M, Perdomo-Celis F, Narváez CF, Salgado DM, Senthoor D, O'Grady M, Phillips E, Durbin A, Fandos D, Miyazaki H, Yen CW, Gélvez-Ramírez M, Warke RV, Ribeiro LS, Teixeira MM, Almeida RP, Muñóz-Medina JE, Ludert JE, Nogueira ML, Colombo TE, Terzian ACB, Bozza PT, Calheiros AS, Vieira YR, Barbosa-Lima G, Vizzoni A, Cerbino-Neto J, Bozza FA, Souza TML, Trugilho MRO, de Filippis AMB, de Sequeira PC, Marques ETA, Magalhaes T, Díaz FJ, Restrepo BN, Marín K, Mattar S, Olson D, Asturias EJ, Lucera M, Singla M, Medigeshi GR, de Bosch N, Tam J, Gómez-Márquez J, Clavet C, Villar L, Hamad-Schifferli K, Gehrke L. Rapid antigen tests for dengue virus serotypes and Zika virus in patient serum. Sci Transl Med 2018; 9:9/409/eaan1589. [PMID: 28954927 DOI: 10.1126/scitranslmed.aan1589] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 06/02/2017] [Accepted: 09/08/2017] [Indexed: 12/16/2022]
Abstract
The recent Zika virus (ZIKV) outbreak demonstrates that cost-effective clinical diagnostics are urgently needed to detect and distinguish viral infections to improve patient care. Unlike dengue virus (DENV), ZIKV infections during pregnancy correlate with severe birth defects, including microcephaly and neurological disorders. Because ZIKV and DENV are related flaviviruses, their homologous proteins and nucleic acids can cause cross-reactions and false-positive results in molecular, antigenic, and serologic diagnostics. We report the characterization of monoclonal antibody pairs that have been translated into rapid immunochromatography tests to specifically detect the viral nonstructural 1 (NS1) protein antigen and distinguish the four DENV serotypes (DENV1-4) and ZIKV without cross-reaction. To complement visual test analysis and remove user subjectivity in reading test results, we used image processing and data analysis for data capture and test result quantification. Using a 30-μl serum sample, the sensitivity and specificity values of the DENV1-4 tests and the pan-DENV test, which detects all four dengue serotypes, ranged from 0.76 to 1.00. Sensitivity/specificity for the ZIKV rapid test was 0.81/0.86, respectively, using a 150-μl serum input. Serum ZIKV NS1 protein concentrations were about 10-fold lower than corresponding DENV NS1 concentrations in infected patients; moreover, ZIKV NS1 protein was not detected in polymerase chain reaction-positive patient urine samples. Our rapid immunochromatography approach and reagents have immediate application in differential clinical diagnosis of acute ZIKV and DENV cases, and the platform can be applied toward developing rapid antigen diagnostics for emerging viruses.
Collapse
Affiliation(s)
- Irene Bosch
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Helena de Puig
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Megan Hiley
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Marc Carré-Camps
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Institut Químic de Sarrià, Universitat Ramon Llull, Barcelona, Spain
| | | | - Carlos F Narváez
- Programa de Medicina, Facultad de Salud, Universidad Surcolombiana, Neiva, Colombia
| | - Doris M Salgado
- Programa de Medicina, Facultad de Salud, Universidad Surcolombiana, Neiva, Colombia
| | - Dewahar Senthoor
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Madeline O'Grady
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Elizabeth Phillips
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ann Durbin
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Program in Virology, Division of Medical Sciences, Harvard Medical School, Boston, MA 02115, USA
| | - Diana Fandos
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Institut Químic de Sarrià, Universitat Ramon Llull, Barcelona, Spain
| | - Hikaru Miyazaki
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Chun-Wan Yen
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Margarita Gélvez-Ramírez
- Universidad Industrial de Santander and AEDES Program (Alianza para el desarrollo de estrategias que disminuyan el impacto de enfermedades transmitidas por Aedes como resultado del estudio de sus endemias y epidemias), Bucaramanga, Santander, Colombia
| | | | - Lucas S Ribeiro
- Immunopharmacology Group, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, Belo Horizonte, Brazil
| | - Mauro M Teixeira
- Immunopharmacology Group, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, Belo Horizonte, Brazil
| | - Roque P Almeida
- Departamento de Medicina Interna e Patologia, Hospital Universitário/Empresa Brasileira de Serviços Hospitalares (EBSERH), Universidade Federal de Sergipe, Aracaju, Brazil
| | - José E Muñóz-Medina
- Laboratorio Central de Epidemiología, Instituto Mexicano del Seguro Social, Avenida Jacarandas S/N, Esquina Circuito Interior, Colonia La Raza Del Azcapotzalco, Código Postal 02990 México D.F., México
| | - Juan E Ludert
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México, México
| | - Mauricio L Nogueira
- Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto, Brazil
| | - Tatiana E Colombo
- Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto, Brazil
| | - Ana C B Terzian
- Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto, Brazil
| | - Patricia T Bozza
- Immunopharmacology Laboratory, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Andrea S Calheiros
- Immunopharmacology Laboratory, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Yasmine R Vieira
- National Institute of Infectious Disease Evandro Chagas, FIOCRUZ, Rio de Janeiro, Brazil
| | - Giselle Barbosa-Lima
- National Institute of Infectious Disease Evandro Chagas, FIOCRUZ, Rio de Janeiro, Brazil
| | - Alexandre Vizzoni
- National Institute of Infectious Disease Evandro Chagas, FIOCRUZ, Rio de Janeiro, Brazil
| | - José Cerbino-Neto
- National Institute of Infectious Disease Evandro Chagas, FIOCRUZ, Rio de Janeiro, Brazil
| | - Fernando A Bozza
- National Institute of Infectious Disease Evandro Chagas, FIOCRUZ, Rio de Janeiro, Brazil.,D'Or Institute of Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Thiago M L Souza
- Immunopharmacology Laboratory, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil.,National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), FIOCRUZ, Rio de Janeiro, Brazil
| | - Monique R O Trugilho
- Toxinology Laboratory and Center for Technological Development in Health (CDTS), FIOCRUZ, Rio de Janeiro, Brazil
| | | | | | - Ernesto T A Marques
- Aggeu Magalhães Research Center, FIOCRUZ, Pernambuco, Recife, Brazil.,Department of Infectious Disease and Microbiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tereza Magalhaes
- Aggeu Magalhães Research Center, FIOCRUZ, Pernambuco, Recife, Brazil.,Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Francisco J Díaz
- Immunovirology Group, School of Medicine, University of Antioquia, Medellín, Colombia
| | - Berta N Restrepo
- Instituto Colombiano de Medicina Tropical (ICMT), Universidad CES, Sabaneta, Antioquia, Colombia
| | - Katerine Marín
- Instituto Colombiano de Medicina Tropical (ICMT), Universidad CES, Sabaneta, Antioquia, Colombia
| | - Salim Mattar
- Universidad de Córdoba, Montería, Córdoba, Colombia
| | - Daniel Olson
- Division of Infectious Diseases, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Edwin J Asturias
- Division of Infectious Diseases, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Mark Lucera
- Division of Infectious Diseases, Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Mohit Singla
- Department of Paediatrics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | | | | | - Justina Tam
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Winchester Engineering Analytical Center (WEAC), Winchester, MA 01890, USA
| | - Jose Gómez-Márquez
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Charles Clavet
- Winchester Engineering Analytical Center (WEAC), Winchester, MA 01890, USA
| | - Luis Villar
- Universidad Industrial de Santander and AEDES Program (Alianza para el desarrollo de estrategias que disminuyan el impacto de enfermedades transmitidas por Aedes como resultado del estudio de sus endemias y epidemias), Bucaramanga, Santander, Colombia
| | - Kimberly Hamad-Schifferli
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. .,Department of Engineering, University of Massachusetts Boston, Boston, MA 02125, USA
| | - Lee Gehrke
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. .,Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
136
|
da Silva LRC. Zika Virus Trafficking and Interactions in the Human Male Reproductive Tract. Pathogens 2018; 7:E51. [PMID: 29751638 PMCID: PMC6027493 DOI: 10.3390/pathogens7020051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/01/2018] [Accepted: 05/02/2018] [Indexed: 12/28/2022] Open
Abstract
Sexual transmission of Zika virus (ZIKV) is a matter of great concern. Infectious viral particles can be shed in semen for as long as six months after infection and can be transferred to male and female sexual partners during unprotected sexual intercourse. The virus can be found inside spermatozoa and could be directly transferred to the oocyte during fertilization. Sexual transmission of ZIKV can contribute to the rise in number of infected individuals in endemic areas as well as in countries where the mosquito vector does not thrive. There is also the possibility, as has been demonstrated in mouse models, that the vaginal deposition of ZIKV particles present in semen could lead to congenital syndrome. In this paper, we review the current literature to understand ZIKV trafficking from the bloodstream to the human male reproductive tract and viral interactions with host cells in interstitial spaces, tubule walls, annexed glands and semen. We hope to highlight gaps to be filled by future research and potential routes for vaccine and antiviral development.
Collapse
|
137
|
Rayner JO, Kalkeri R, Goebel S, Cai Z, Green B, Lin S, Snyder B, Hagelin K, Walters KB, Koide F. Comparative Pathogenesis of Asian and African-Lineage Zika Virus in Indian Rhesus Macaque's and Development of a Non-Human Primate Model Suitable for the Evaluation of New Drugs and Vaccines. Viruses 2018; 10:E229. [PMID: 29723973 PMCID: PMC5977222 DOI: 10.3390/v10050229] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 04/24/2018] [Accepted: 04/27/2018] [Indexed: 11/24/2022] Open
Abstract
The establishment of a well characterized non-human primate model of Zika virus (ZIKV) infection is critical for the development of medical interventions. In this study, challenging Indian rhesus macaques (IRMs) with ZIKV strains of the Asian lineage resulted in dose-dependent peak viral loads between days 2 and 5 post infection and a robust immune response which protected the animals from homologous and heterologous re-challenge. In contrast, viremia in IRMs challenged with an African lineage strain was below the assay’s lower limit of quantitation, and the immune response was insufficient to protect from re-challenge. These results corroborate previous observations but are contrary to reports using other African strains, obviating the need for additional studies to elucidate the variables contributing to the disparities. Nonetheless, the utility of an Asian lineage ZIKV IRM model for countermeasure development was verified by vaccinating animals with a formalin inactivated reference vaccine and demonstrating sterilizing immunity against a subsequent subcutaneous challenge.
Collapse
Affiliation(s)
- Jonathan O Rayner
- Department of Infectious Disease Research, Southern Research Institute, Birmingham, AL 35205, USA.
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL 36688, USA.
| | - Raj Kalkeri
- Department of Infectious Disease Research, Southern Research Institute, Frederick, MD 21701, USA.
| | - Scott Goebel
- Department of Infectious Disease Research, Southern Research Institute, Frederick, MD 21701, USA.
| | - Zhaohui Cai
- Department of Infectious Disease Research, Southern Research Institute, Frederick, MD 21701, USA.
| | - Brian Green
- Department of Infectious Disease Research, Southern Research Institute, Frederick, MD 21701, USA.
| | - Shuling Lin
- Department of Infectious Disease Research, Southern Research Institute, Frederick, MD 21701, USA.
| | - Beth Snyder
- Department of Infectious Disease Research, Southern Research Institute, Frederick, MD 21701, USA.
| | - Kimberly Hagelin
- Department of Infectious Disease Research, Southern Research Institute, Frederick, MD 21701, USA.
| | - Kevin B Walters
- Department of Infectious Disease Research, Southern Research Institute, Frederick, MD 21701, USA.
| | - Fusataka Koide
- Department of Infectious Disease Research, Southern Research Institute, Frederick, MD 21701, USA.
| |
Collapse
|
138
|
Stassen L, Armitage CW, van der Heide DJ, Beagley KW, Frentiu FD. Zika Virus in the Male Reproductive Tract. Viruses 2018; 10:v10040198. [PMID: 29659541 PMCID: PMC5923492 DOI: 10.3390/v10040198] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/11/2018] [Accepted: 04/13/2018] [Indexed: 01/07/2023] Open
Abstract
Arthropod-borne viruses (arboviruses) are resurging across the globe. Zika virus (ZIKV) has caused significant concern in recent years because it can lead to congenital malformations in babies and Guillain-Barré syndrome in adults. Unlike other arboviruses, ZIKV can be sexually transmitted and may persist in the male reproductive tract. There is limited information regarding the impact of ZIKV on male reproductive health and fertility. Understanding the mechanisms that underlie persistent ZIKV infections in men is critical to developing effective vaccines and therapies. Mouse and macaque models have begun to unravel the pathogenesis of ZIKV infection in the male reproductive tract, with the testes and prostate gland implicated as potential reservoirs for persistent ZIKV infection. Here, we summarize current knowledge regarding the pathogenesis of ZIKV in the male reproductive tract, the development of animal models to study ZIKV infection at this site, and prospects for vaccines and therapeutics against persistent ZIKV infection.
Collapse
Affiliation(s)
- Liesel Stassen
- Institute of Health and Biomedical Innovation, and School of Biomedical Sciences, Queensland University of Technology, Brisbane 4006, Queensland, Australia.
| | - Charles W Armitage
- Institute of Health and Biomedical Innovation, and School of Biomedical Sciences, Queensland University of Technology, Brisbane 4006, Queensland, Australia.
| | - David J van der Heide
- Institute of Health and Biomedical Innovation, and School of Biomedical Sciences, Queensland University of Technology, Brisbane 4006, Queensland, Australia.
| | - Kenneth W Beagley
- Institute of Health and Biomedical Innovation, and School of Biomedical Sciences, Queensland University of Technology, Brisbane 4006, Queensland, Australia.
| | - Francesca D Frentiu
- Institute of Health and Biomedical Innovation, and School of Biomedical Sciences, Queensland University of Technology, Brisbane 4006, Queensland, Australia.
| |
Collapse
|
139
|
Mavigner M, Raper J, Kovacs-Balint Z, Gumber S, O'Neal JT, Bhaumik SK, Zhang X, Habib J, Mattingly C, McDonald CE, Avanzato V, Burke MW, Magnani DM, Bailey VK, Watkins DI, Vanderford TH, Fair D, Earl E, Feczko E, Styner M, Jean SM, Cohen JK, Silvestri G, Johnson RP, O'Connor DH, Wrammert J, Suthar MS, Sanchez MM, Alvarado MC, Chahroudi A. Postnatal Zika virus infection is associated with persistent abnormalities in brain structure, function, and behavior in infant macaques. Sci Transl Med 2018; 10:eaao6975. [PMID: 29618564 PMCID: PMC6186170 DOI: 10.1126/scitranslmed.aao6975] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 03/07/2018] [Indexed: 12/22/2022]
Abstract
The Zika virus (ZIKV) epidemic is associated with fetal brain lesions and other serious birth defects classified as congenital ZIKV syndrome. Postnatal ZIKV infection in infants and children has been reported; however, data on brain anatomy, function, and behavioral outcomes following infection are absent. We show that postnatal ZIKV infection of infant rhesus macaques (RMs) results in persistent structural and functional alterations of the central nervous system compared to age-matched controls. We demonstrate ZIKV lymphoid tropism and neurotropism in infant RMs and histopathologic abnormalities in the peripheral and central nervous systems including inflammatory infiltrates, astrogliosis, and Wallerian degeneration. Structural and resting-state functional magnetic resonance imaging (MRI/rs-fMRI) show persistent enlargement of lateral ventricles, maturational changes in specific brain regions, and altered functional connectivity (FC) between brain areas involved in emotional behavior and arousal functions, including weakened amygdala-hippocampal connectivity in two of two ZIKV-infected infant RMs several months after clearance of ZIKV RNA from peripheral blood. ZIKV infection also results in distinct alterations in the species-typical emotional reactivity to acute stress, which were predicted by the weak amygdala-hippocampal FC. We demonstrate that postnatal ZIKV infection of infants in this model affects neurodevelopment, suggesting that long-term clinical monitoring of pediatric cases is warranted.
Collapse
Affiliation(s)
- Maud Mavigner
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jessica Raper
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Zsofia Kovacs-Balint
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Sanjeev Gumber
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | | | - Siddhartha K Bhaumik
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Xiaodong Zhang
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Jakob Habib
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Cameron Mattingly
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | - Victoria Avanzato
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Mark W Burke
- Department of Physiology and Biophysics, Howard University, Washington, DC 20060, USA
| | - Diogo M Magnani
- Department of Pathology, University of Miami, Miami, FL 33146, USA
| | - Varian K Bailey
- Department of Pathology, University of Miami, Miami, FL 33146, USA
| | - David I Watkins
- Department of Pathology, University of Miami, Miami, FL 33146, USA
| | - Thomas H Vanderford
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Damien Fair
- Oregon Health and Science University, Portland, OR 97239, USA
| | - Eric Earl
- Oregon Health and Science University, Portland, OR 97239, USA
| | - Eric Feczko
- Oregon Health and Science University, Portland, OR 97239, USA
| | - Martin Styner
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Sherrie M Jean
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Joyce K Cohen
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Guido Silvestri
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - R Paul Johnson
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - David H O'Connor
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Jens Wrammert
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Mehul S Suthar
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Emory Vaccine Center, Atlanta, GA 30329, USA
| | - Mar M Sanchez
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Maria C Alvarado
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA.
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| |
Collapse
|
140
|
Kumar A, Jovel J, Lopez-Orozco J, Limonta D, Airo AM, Hou S, Stryapunina I, Fibke C, Moore RB, Hobman TC. Human Sertoli cells support high levels of Zika virus replication and persistence. Sci Rep 2018; 8:5477. [PMID: 29615760 PMCID: PMC5883016 DOI: 10.1038/s41598-018-23899-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 03/20/2018] [Indexed: 02/07/2023] Open
Abstract
Zika virus is a teratogenic mosquito-transmitted flavivirus that is associated with birth defects in newborns and Guillain–Barré syndrome in adults. The virus can also be sexually transmitted, but currently, very little is known about the cell types supporting virus replication and persistence in human testes. Using primary cell cultures, we observed that Sertoli but not Leydig cells are highly susceptible to Zika virus infection, a process that is dependent on the TAM family receptor Axl. In cell culture, Sertoli cells could be productively infected with Zika virus for at least 6-weeks. Infection of Sertoli cells resulted in dramatic changes to the transcriptional profile of these cells. The most upregulated mRNA in infected cells was basic fibroblast growth factor (FGF2), a cytokine that was found to enhance Zika virus replication and support viral persistence. Together these findings provide key insights into understanding how Zika virus persists in the male reproductive tract and in turn may aid in developing antiviral therapies or strategies to minimize sexual transmission of this pathogen.
Collapse
Affiliation(s)
- Anil Kumar
- Departments of Cell Biology, University of Alberta, Edmonton, Canada
| | - Juan Jovel
- Departments of Medicine, University of Alberta, Edmonton, Canada
| | | | - Daniel Limonta
- Departments of Cell Biology, University of Alberta, Edmonton, Canada
| | - Adriana M Airo
- Departments of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada
| | - Shangmei Hou
- Departments of Cell Biology, University of Alberta, Edmonton, Canada
| | - Iryna Stryapunina
- Departments of Cell Biology, University of Alberta, Edmonton, Canada
| | - Chad Fibke
- Departments of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada
| | - Ronald B Moore
- Departments of Surgery, University of Alberta, Edmonton, Canada
| | - Tom C Hobman
- Departments of Cell Biology, University of Alberta, Edmonton, Canada. .,Departments of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada. .,Li Ka Shing Institute of Virology, Edmonton, Canada. .,Women & Children's Health Research Institute, Edmonton, Canada.
| |
Collapse
|
141
|
Abstract
The spread of Zika virus to the Americas was accompanied by surge in the number of infants with CNS abnormalities leading to a declaration of a health emergency by the WHO. This was accompanied by significant responses from governmental health agencies in the United States and Europe that resulted in significant new information described in the natural history of this perinatal infection in a very short period of time. Although much has been learned about Zika virus infection during pregnancy, limitations of current diagnostics and the challenges for accurate serologic diagnosis of acute Zika virus infection has restricted our understanding of the natural history of this perinatal infection to infants born to women with clinical disease during pregnancy and to Zika exposed infants with obvious clinical stigmata of disease. Thus, the spectrum of disease in infants exposed to Zika virus during pregnancy remains to be defined. In contrast, observations in informative animal models of Zika virus infections have provided rational pathways for vaccine development and existing antiviral drug development programs for other flaviviruses have resulted in accelerated development for potential antiviral therapies. This brief review will highlight some of the current concepts of the natural history of Zika virus during pregnancy.
Collapse
Affiliation(s)
- William J Britt
- Department of Pediatrics, University of Alabama School of Medicine, Childrens Hospital Harbor Bldg 160, Birmingham, AL 35233.
| |
Collapse
|
142
|
Duggal NK, McDonald EM, Ritter JM, Brault AC. Sexual transmission of Zika virus enhances in utero transmission in a mouse model. Sci Rep 2018; 8:4510. [PMID: 29540804 PMCID: PMC5852059 DOI: 10.1038/s41598-018-22840-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/28/2018] [Indexed: 12/20/2022] Open
Abstract
Zika virus (ZIKV) is an emerging mosquito-borne virus that can cause ZIKV congenital syndrome when a pregnant woman is infected. Sexual transmission has also been described for ZIKV, though the relationship between sexual transmission and vertical transmission has not been investigated. Here, viral dissemination to the female reproductive tract and fetuses was assessed in immunodeficient (AG129) female mice that were exposed to ZIKV by subcutaneous (s.c.) inoculation, intravaginal (ivag.) inoculation, or sexual transmission from infected male AG129 mice. Pregnant females had significantly increased ZIKV dissemination to the female reproductive tract compared to non-pregnant females when exposed by s.c. or ivag. inoculation. Sexual transmission resulted in significantly greater morbidity and mortality in females and higher ZIKV titers in the female reproductive tract than s.c. or ivag. inoculation. Ovaries from females infected sexually contained ZIKV RNA within the ovarian follicles. Furthermore, ZIKV titers were significantly higher in fetuses from dams exposed sexually compared to fetuses from dams exposed by s.c. or ivag. inoculation. These results demonstrate that sexual transmission enhances dissemination of ZIKV to the female reproductive tract and developing fetuses in a mouse model.
Collapse
Affiliation(s)
- Nisha K Duggal
- Division of Vector-borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, USA
| | - Erin M McDonald
- Division of Vector-borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, USA
| | - Jana M Ritter
- Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Aaron C Brault
- Division of Vector-borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, USA.
| |
Collapse
|
143
|
Cellular and Humoral Immunity Protect against Vaginal Zika Virus Infection in Mice. J Virol 2018; 92:JVI.00038-18. [PMID: 29343577 PMCID: PMC5972878 DOI: 10.1128/jvi.00038-18] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 01/07/2023] Open
Abstract
Zika virus (ZIKV), which can cause devastating disease in fetuses of infected pregnant women, can be transmitted by mosquito inoculation and sexual routes. Little is known about immune protection against sexually transmitted ZIKV. In this study, we show that previous infection through intravaginal or subcutaneous routes with a contemporary Brazilian strain of ZIKV can protect against subsequent intravaginal challenge with a homologous strain. Both routes of inoculation induced high titers of ZIKV-specific and neutralizing antibody in serum and the vaginal lumen. Virus-specific T cells were recruited to and retained in the female reproductive tract after intravaginal and subcutaneous ZIKV infection. Studies in mice with genetic or acquired deficiencies in B and/or T cells demonstrated that both lymphocyte populations redundantly protect against intravaginal challenge in ZIKV-immune animals. Passive transfer of ZIKV-immune IgG or T cells significantly limited intravaginal infection of naive mice, although antibody more effectively prevented dissemination throughout the reproductive tract. Collectively, our experiments begin to establish the immune correlates of protection against intravaginal ZIKV infection, which should inform vaccination strategies in nonpregnant and pregnant women.IMPORTANCE The recent ZIKV epidemic resulted in devastating outcomes in fetuses and may affect reproductive health. Unlike other flaviviruses, ZIKV can be spread by sexual contact as well as a mosquito vector. While previous studies have identified correlates of protection for mosquito-mediated infection, few have focused on immunity against sexual transmission. As exposure to ZIKV via mosquito bite has likely occurred to many living in areas where ZIKV is endemic, our study addresses whether this route of infection can protect against subsequent sexual exposure. We demonstrate that subcutaneous ZIKV infection can protect against subsequent vaginal infection by generating both local antiviral T cell and antibody responses. Our research begins to define the immune correlates of protection for ZIKV infection in the vagina and provides a foundation for testing ZIKV vaccines against sexual transmission.
Collapse
|
144
|
Soriano-Arandes A, Rivero-Calle I, Nastouli E, Espiau M, Frick MA, Alarcon A, Martinón-Torres F. What we know and what we don't know about perinatal Zika virus infection: a systematic review. Expert Rev Anti Infect Ther 2018; 16:243-254. [PMID: 29415586 DOI: 10.1080/14787210.2018.1438265] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Zika virus (ZIKV) infection has caused the most challenging worldwide infectious epidemic outbreak in recent months. ZIKV causes microcephaly and other congenital malformations. There is a need to perform updated systematic reviews on ZIKV infection periodically because this epidemic is bringing up new evidence with extraordinary speed. Areas covered: Evidence related to ZIKV infection in the gestational, perinatal, and early infant periods covering epidemiology, virology, pathogenesis, risk factors, time of infection during pregnancy, newborn symptoms, treatment, and vaccines. To this end, a search was performed using terms ['Zika'] AND ['Perinatal Infection'] OR ['Congenital Infection'] in the PubMed® international electronic database. Out of a total of 1,538 articles published until 30 November 2017, we finally assessed 106 articles articles that were relevant to the research areas included in this study. Expert commentary: ZIKV is a new teratogenic/neurotropic virus affecting fetuses. Many challenges are still far from being solved regarding the epidemiology, case definition, clinical and laboratory diagnosis, and preventive measures. An approach using 'omics' and new biomarkers for diagnosis, and a ZIKV-vaccine for treatment, might finally give us the tools to solve these challenges.
Collapse
Affiliation(s)
- Antoni Soriano-Arandes
- a Pediatric Infectious Diseases and Immunodeficiencies Unit , Hospital Universitari Vall d'Hebron , Barcelona , Spain
| | - Irene Rivero-Calle
- b Translational Pediatrics and Infectious Diseases, Department of Pediatrics , Complejo Hospitalario Universitario de Santiago de Compostela , Santiago de Compostela , Spain
| | - Eleni Nastouli
- c Department of Virology , University College of London Hospitals NHS Foundation Trust , London , UK
| | - Maria Espiau
- a Pediatric Infectious Diseases and Immunodeficiencies Unit , Hospital Universitari Vall d'Hebron , Barcelona , Spain
| | - M A Frick
- a Pediatric Infectious Diseases and Immunodeficiencies Unit , Hospital Universitari Vall d'Hebron , Barcelona , Spain
| | - Ana Alarcon
- d Department of Neonatology , Hospital Universitari Sant Joan de Déu , Barcelona , Spain
| | - Federico Martinón-Torres
- b Translational Pediatrics and Infectious Diseases, Department of Pediatrics , Complejo Hospitalario Universitario de Santiago de Compostela , Santiago de Compostela , Spain
| |
Collapse
|
145
|
Spencer JL, Lahon A, Tran LL, Arya RP, Kneubehl AR, Vogt MB, Xavier D, Rowley DR, Kimata JT, Rico-Hesse RR. Replication of Zika Virus in Human Prostate Cells: A Potential Source of Sexually Transmitted Virus. J Infect Dis 2018; 217:538-547. [PMID: 28968863 PMCID: PMC5853941 DOI: 10.1093/infdis/jix436] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 08/21/2017] [Indexed: 12/28/2022] Open
Abstract
Background While Zika virus (ZIKV) is mainly transmitted by mosquitoes, numerous cases of sexual transmission have been reported during recent outbreaks. Little is known about which host cell types or entry factors aid in mediating this sexual transmission. Methods In this study, we investigated ZIKV cell tropism by infecting 2 types of human prostate cells with 3 contemporary ZIKV isolates from persons infected in the Americas. We used real-time quantitative polymerase chain reaction and immunofluorescence analyses to measure infection and flow cytometry to detect entry factor expression. Results Here we show that ZIKV infects, replicates, and produces infectious virus in prostate stromal mesenchymal stem cells, epithelial cells, and organoids made with a combination of these cells. We also show that prostate cells express several well-characterized flavivirus attachment factors. In contrast, dengue virus does not infect or does not replicate in these prostate cells, although it is known to use similar receptors. Conclusions Our results indicate that ZIKV favors infection of stromal cells more so than epithelial cells in organoids, possibly indicating a preference for stem cells in general. Overall, these results suggest that ZIKV replication occurs in the human prostate and can account for ZIKV secretion in semen, thus leading to sexual transmission.
Collapse
Affiliation(s)
| | | | - Linda L Tran
- Department of Molecular and Cellular Biology
- Integrative Molecular and Biomedical Sciences Program
| | - Ravi P Arya
- Department of Molecular Virology and Microbiology
| | | | - Megan B Vogt
- Department of Molecular Virology and Microbiology
- Integrative Molecular and Biomedical Sciences Program
| | - Daniela Xavier
- Genomic and RNA Profiling Core, Baylor College of Medicine, Houston, Texas
| | | | | | - Rebecca R Rico-Hesse
- Department of Molecular Virology and Microbiology
- Correspondence: R. R. Rico-Hesse, MPH, PhD, Department of Molecular Virology and Microbiology, Baylor College of Medicine, 1 Baylor Plaza, MS 385, Houston, TX 77030 ()
| |
Collapse
|
146
|
Development of a chimeric Zika vaccine using a licensed live-attenuated flavivirus vaccine as backbone. Nat Commun 2018; 9:673. [PMID: 29445153 PMCID: PMC5813210 DOI: 10.1038/s41467-018-02975-w] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 01/10/2018] [Indexed: 01/07/2023] Open
Abstract
The global spread of Zika virus (ZIKV) and its unexpected association with congenital defects necessitates the rapid development of a safe and effective vaccine. Here we report the development and characterization of a recombinant chimeric ZIKV vaccine candidate (termed ChinZIKV) that expresses the prM-E proteins of ZIKV using the licensed Japanese encephalitis live-attenuated vaccine SA14-14-2 as the genetic backbone. ChinZIKV retains its replication activity and genetic stability in vitro, while exhibiting an attenuation phenotype in multiple animal models. Remarkably, immunization of mice and rhesus macaques with a single dose of ChinZIKV elicits robust and long-lasting immune responses, and confers complete protection against ZIKV challenge. Significantly, female mice immunized with ChinZIKV are protected against placental and fetal damage upon ZIKV challenge during pregnancy. Overall, our study provides an alternative vaccine platform in response to the ZIKV emergency, and the safety, immunogenicity, and protection profiles of ChinZIKV warrant further clinical development.
Collapse
|
147
|
Zmurko J, Vasey DB, Donald CL, Armstrong AA, McKee ML, Kohl A, Clayton RF. Mitigating the risk of Zika virus contamination of raw materials and cell lines in the manufacture of biologicals. J Gen Virol 2018; 99:219-229. [PMID: 29239715 PMCID: PMC5882083 DOI: 10.1099/jgv.0.000995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/04/2017] [Indexed: 01/14/2023] Open
Abstract
Ensuring the virological safety of biologicals is challenging due to the risk of viral contamination of raw materials and cell banks, and exposure during in-process handling to known and/or emerging viral pathogens. Viruses may contaminate raw materials and biologicals intended for human or veterinary use and remain undetected until appropriate testing measures are employed. The outbreak and expansive spread of the mosquito-borne flavivirus Zika virus (ZIKV) poses challenges to screening human- and animal -derived products used in the manufacture of biologicals. Here, we report the results of an in vitro study where detector cell lines were challenged with African and Asian lineages of ZIKV. We demonstrate that this pathogen is robustly detectable by in vitro assay, thereby providing assurance of detection of ZIKV, and in turn underpinning the robustness of in vitro virology assays in safety testing of biologicals.
Collapse
Affiliation(s)
- Joanna Zmurko
- Merck KGaA, BioReliance® Services, Todd Campus, West of Scotland Science Park, Glasgow G20 OXA, UK
| | - Douglas B. Vasey
- Merck KGaA, BioReliance® Services, Todd Campus, West of Scotland Science Park, Glasgow G20 OXA, UK
| | - Claire L. Donald
- MRC-University of Glasgow Centre for Virus Research, Glasgow G61 1QH, UK
| | - Alison A. Armstrong
- Merck KGaA, BioReliance® Services, Todd Campus, West of Scotland Science Park, Glasgow G20 OXA, UK
| | - Marian L. McKee
- Merck KGaA, BioReliance® Services, Todd Campus, West of Scotland Science Park, Glasgow G20 OXA, UK
| | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, Glasgow G61 1QH, UK
| | - Reginald F. Clayton
- Merck KGaA, BioReliance® Services, Todd Campus, West of Scotland Science Park, Glasgow G20 OXA, UK
| |
Collapse
|
148
|
Mohr EL, Block LN, Newman CM, Stewart LM, Koenig M, Semler M, Breitbach ME, Teixeira LBC, Zeng X, Weiler AM, Barry GL, Thoong TH, Wiepz GJ, Dudley DM, Simmons HA, Mejia A, Morgan TK, Salamat MS, Kohn S, Antony KM, Aliota MT, Mohns MS, Hayes JM, Schultz-Darken N, Schotzko ML, Peterson E, Capuano S, Osorio JE, O’Connor SL, Friedrich TC, O’Connor DH, Golos TG. Ocular and uteroplacental pathology in a macaque pregnancy with congenital Zika virus infection. PLoS One 2018; 13:e0190617. [PMID: 29381706 PMCID: PMC5790226 DOI: 10.1371/journal.pone.0190617] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/18/2017] [Indexed: 12/03/2022] Open
Abstract
Congenital Zika virus (ZIKV) infection impacts fetal development and pregnancy outcomes. We infected a pregnant rhesus macaque with a Puerto Rican ZIKV isolate in the first trimester. The pregnancy was complicated by preterm premature rupture of membranes (PPROM), intraamniotic bacterial infection and fetal demise 49 days post infection (gestational day 95). Significant pathology at the maternal-fetal interface included acute chorioamnionitis, placental infarcts, and leukocytoclastic vasculitis of the myometrial radial arteries. ZIKV RNA was disseminated throughout fetal tissues and maternal immune system tissues at necropsy, as assessed by quantitative RT-PCR for viral RNA. Replicating ZIKV was identified in fetal tissues, maternal uterus, and maternal spleen by fluorescent in situ hybridization for viral replication intermediates. Fetal ocular pathology included a choroidal coloboma, suspected anterior segment dysgenesis, and a dysplastic retina. This is the first report of ocular pathology and prolonged viral replication in both maternal and fetal tissues following congenital ZIKV infection in a rhesus macaque. PPROM followed by fetal demise and severe pathology of the visual system have not been described in macaque congenital ZIKV infection previously. While this case of ZIKV infection during pregnancy was complicated by bacterial infection with PPROM, the role of ZIKV on this outcome cannot be precisely defined, and further nonhuman primate studies will determine if increased risk for PPROM or other adverse pregnancy outcomes are associated with congenital ZIKV infection.
Collapse
Affiliation(s)
- Emma L. Mohr
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail: (ELM); (TGG)
| | - Lindsey N. Block
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Christina M. Newman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Laurel M. Stewart
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Michelle Koenig
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Matthew Semler
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Meghan E. Breitbach
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Leandro B. C. Teixeira
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Xiankun Zeng
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Andrea M. Weiler
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Gabrielle L. Barry
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Troy H. Thoong
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Gregory J. Wiepz
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Dawn M. Dudley
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Heather A. Simmons
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Andres Mejia
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Terry K. Morgan
- Departments of Pathology and Obstetrics & Gynecology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - M. Shahriar Salamat
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Sarah Kohn
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Kathleen M. Antony
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Matthew T. Aliota
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Mariel S. Mohns
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jennifer M. Hayes
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Nancy Schultz-Darken
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Michele L. Schotzko
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Eric Peterson
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Saverio Capuano
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jorge E. Osorio
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Shelby L. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Thomas C. Friedrich
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - David H. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Thaddeus G. Golos
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail: (ELM); (TGG)
| |
Collapse
|
149
|
Abstract
Flaviviruses such as dengue (DENV), yellow fever (YFV), West Nile (WNV), and Zika (ZIKV) are human pathogens of global significance. In particular, DENV causes the most prevalent mosquito-borne viral diseases in humans, and ZIKV emerged from obscurity into the spotlight in 2016 as the etiologic agent of congenital Zika syndrome. Owing to the recent emergence of ZIKV as a global pandemic threat, the roles of the immune system during ZIKV infections are as yet unclear. In contrast, decades of DENV research implicate a dual role for the immune system in protection against and pathogenesis of DENV infection. As DENV and ZIKV are closely related, knowledge based on DENV studies has been used to prioritize investigation of ZIKV immunity and pathogenesis, and to accelerate ZIKV diagnostic, therapeutic, and vaccine design. This review discusses the following topics related to innate and adaptive immune responses to DENV and ZIKV: the interferon system as the key mechanism of host defense and viral target for immune evasion, antibody-mediated protection versus antibody-dependent enhancement, and T cell-mediated protection versus original T cell antigenic sin. Understanding the mechanisms that regulate the balance between immune-mediated protection and pathogenesis during DENV and ZIKV infections is critical toward development of safe and effective DENV and ZIKV therapeutics and vaccines.
Collapse
Affiliation(s)
- Annie Elong Ngono
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA;
| | - Sujan Shresta
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA;
| |
Collapse
|
150
|
Zika virus infection in pregnant rhesus macaques causes placental dysfunction and immunopathology. Nat Commun 2018; 9:263. [PMID: 29343712 PMCID: PMC5772047 DOI: 10.1038/s41467-017-02499-9] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 12/04/2017] [Indexed: 12/13/2022] Open
Abstract
Zika virus (ZIKV) infection during pregnancy leads to an increased risk of fetal growth restriction and fetal central nervous system malformations, which are outcomes broadly referred to as the Congenital Zika Syndrome (CZS). Here we infect pregnant rhesus macaques and investigate the impact of persistent ZIKV infection on uteroplacental pathology, blood flow, and fetal growth and development. Despite seemingly normal fetal growth and persistent fetal-placenta-maternal infection, advanced non-invasive in vivo imaging studies reveal dramatic effects on placental oxygen reserve accompanied by significantly decreased oxygen permeability of the placental villi. The observation of abnormal oxygen transport within the placenta appears to be a consequence of uterine vasculitis and placental villous damage in ZIKV cases. In addition, we demonstrate a robust maternal-placental-fetal inflammatory response following ZIKV infection. This animal model reveals a potential relationship between ZIKV infection and uteroplacental pathology that appears to affect oxygen delivery to the fetus during development. Zika virus infection during pregnancy can result in birth defects, but underlying pathogenesis at the maternal-fetal interface is unclear. Here, the authors use non-invasive in vivo imaging of Zika-infected rhesus macaques and show that infection results in abnormal oxygen transport across the placenta.
Collapse
|