1
|
Wessel RE, Dolatshahi S. Regulators of placental antibody transfer through a modeling lens. Nat Immunol 2024; 25:2024-2036. [PMID: 39379658 DOI: 10.1038/s41590-024-01971-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/03/2024] [Indexed: 10/10/2024]
Abstract
Infants are vulnerable to infections owing to a limited ability to mount a humoral immune response and their tolerogenic immune phenotype, which has impeded the success of newborn vaccination. Transplacental transfer of IgG from mother to fetus provides crucial protection in the first weeks of life, and maternal immunization has recently been implemented as a public health strategy to protect newborns against serious infections. Despite their early success, current maternal vaccines do not provide comparable protection across pregnancies with varying gestational lengths and placental and maternal immune features, and they do not account for the dynamic interplay between the maternal immune response and placental transfer. Moreover, progress toward the rational design of maternal vaccines has been hindered by inadequacies of existing experimental models and safety challenges of investigating longitudinal dynamics of IgG transfer in pregnant humans. Alternatively, in silico mechanistic models are a logical framework to disentangle the processes regulating placental antibody transfer. This Review synthesizes current literature through a mechanistic modeling lens to identify placental and maternal regulators of antibody transfer, their clinical covariates, and knowledge gaps to guide future research. We also describe opportunities to use integrated modeling and experimental approaches toward the rational design of vaccines against existing and emerging neonatal pathogen threats.
Collapse
Affiliation(s)
- Remziye E Wessel
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Virginia, Charlottesville, VA, USA
| | - Sepideh Dolatshahi
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Virginia, Charlottesville, VA, USA.
- Carter Immunology Center, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
2
|
da Costa Castilho M, de Filippis AMB, Machado LC, de Lima Calvanti TYV, Lima MC, Fonseca V, Giovanetti M, Docena C, Neto AM, Bôtto-Menezes CHA, Kara EO, de La Barrera R, Modjarrad K, Giozza SP, Pereira GF, Alcantara LCJ, Broutet NJN, Calvet GA, Wallau GL, Franca RFO. Evidence of Zika Virus Reinfection by Genome Diversity and Antibody Response Analysis, Brazil. Emerg Infect Dis 2024; 30:310-320. [PMID: 38270216 PMCID: PMC10826783 DOI: 10.3201/eid3002.230122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024] Open
Abstract
We generated 238 Zika virus (ZIKV) genomes from 135 persons in Brazil who had samples collected over 1 year to evaluate virus persistence. Phylogenetic inference clustered the genomes together with previously reported ZIKV strains from northern Brazil, showing that ZIKV has been remained relatively stable over time. Temporal phylogenetic analysis revealed limited within-host diversity among most ZIKV-persistent infected associated samples. However, we detected unusual virus temporal diversity from >5 persons, uncovering the existence of divergent genomes within the same patient. All those patients showed an increase in neutralizing antibody levels, followed by a decline at the convalescent phase of ZIKV infection. Of interest, in 3 of those patients, titers of neutralizing antibodies increased again after 6 months of ZIKV infection, concomitantly with real-time reverse transcription PCR re-positivity, supporting ZIKV reinfection events. Altogether, our findings provide evidence for the existence of ZIKV reinfection events.
Collapse
|
3
|
Nogales A, Martínez-Sobrido L, Almazán F. Reverse Genetics of Zika Virus Using a Bacterial Artificial Chromosome. Methods Mol Biol 2024; 2733:185-206. [PMID: 38064034 DOI: 10.1007/978-1-0716-3533-9_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Zika virus (ZIKV) is a mosquito-borne member of the Flaviviridae family that has become a global threat to human health. Although ZIKV has been known to circulate for decades causing mild febrile illness, the more recent ZIKV outbreaks in the Americas and the Caribbean have been associated with severe neurological disorders and congenital abnormalities. The development of ZIKV reverse genetics approaches have allowed researchers to address key questions on the biology of ZIKV by genetically engineering infectious recombinant (r)ZIKV. This has resulted in a better understanding of the biology of ZIKV infections, including viral pathogenesis, molecular mechanisms of viral replication and transcription, or the interaction of viral and host factors, among others aspects. In addition, reverse genetics systems have facilitated the identification of anti-ZIKV compounds and the development of new prophylactic approaches to combat ZIKV infections. Different reverse genetics strategies have been implemented for the recovery of rZIKV. All these reverse genetics systems have faced and overcome multiple challenges, including the viral genome size, the toxicity of viral sequences in bacteria, etc. In this chapter we describe the generation of a ZIKV full-length complementary (c)DNA infectious clone based on the use of a bacterial artificial chromosome (BAC) and the experimental procedures for the successful recovery of rZIKV. Importantly, the protocol described in this chapter provides a powerful method for the generation of infectious clones of other flaviviruses with genomes that have stability problems during bacterial propagation.
Collapse
Affiliation(s)
- Aitor Nogales
- Centro de Investigación en Sanidad Animal (CISA-INIA/CSIC), Madrid, Spain
| | | | - Fernando Almazán
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB), CSIC, Madrid, Spain.
| |
Collapse
|
4
|
Jain S, Vimal N, Angmo N, Sengupta M, Thangaraj S. Dengue Vaccination: Towards a New Dawn of Curbing Dengue Infection. Immunol Invest 2023; 52:1096-1149. [PMID: 37962036 DOI: 10.1080/08820139.2023.2280698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Dengue is an infectious disease caused by dengue virus (DENV) and is a serious global burden. Antibody-dependent enhancement and the ability of DENV to infect immune cells, along with other factors, lead to fatal Dengue Haemorrhagic Fever and Dengue Shock Syndrome. This necessitates the development of a robust and efficient vaccine but vaccine development faces a number of hurdles. In this review, we look at the epidemiology, genome structure and cellular targets of DENV and elaborate upon the immune responses generated by human immune system against DENV infection. The review further sheds light on various challenges in development of a potent vaccine against DENV which is followed by presenting a current account of different vaccines which are being developed or have been licensed.
Collapse
Affiliation(s)
- Sidhant Jain
- Independent Researcher, Institute for Globally Distributed Open Research and Education (IGDORE), Rewari, India
| | - Neha Vimal
- Bhaskaracharya College of Applied Sciences, University of Delhi, Delhi, India
| | - Nilza Angmo
- Maitreyi College, University of Delhi, Delhi, India
| | - Madhumita Sengupta
- Janki Devi Bajaj Government Girls College, University of Kota, Kota, India
| | - Suraj Thangaraj
- Swami Ramanand Teerth Rural Government Medical College, Maharashtra University of Health Sciences, Ambajogai, India
| |
Collapse
|
5
|
Saron WAA, Shanmugam K, Tung CC, Patmanathan RK, Rathore APS, Anderson DE, St John AL. Exacerbated Zika virus-induced neuropathology and microcephaly in fetuses of dengue-immune nonhuman primates. Sci Transl Med 2023; 15:eadd2420. [PMID: 37878671 DOI: 10.1126/scitranslmed.add2420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 10/04/2023] [Indexed: 10/27/2023]
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus that can vertically transmit from mother to fetus, potentially causing congenital defects, including microcephaly. It is not fully understood why some fetuses experience severe complications after in utero exposure to ZIKV, whereas others do not. Given the antigenic similarity between ZIKV and the closely related virus dengue (DENV) and the potential of DENV-specific antibodies to enhance ZIKV disease severity in mice, we questioned whether maternal DENV immunity could influence fetal outcomes in a nonhuman primate model of ZIKV vertical transmission. We found significantly increased severity of congenital Zika syndrome (CZS) in fetuses of DENV-immune cynomolgus macaques infected with ZIKV in early pregnancy compared with naïve controls, which occurred despite no effect on maternal ZIKV infection or antibody responses. Ultrasound measurements of head circumference and biparietal diameter measurements taken sequentially throughout pregnancy demonstrated CZS in fetuses of DENV-immune pregnant macaques. Furthermore, severe CZS enhanced by DENV immunity was typified by reduced cortical thickness and increased frequency of neuronal death, hemorrhaging, cellular infiltrations, calcifications, and lissencephaly in fetal brains. This study shows that maternal immunity to DENV can worsen ZIKV neurological outcomes in fetal primates, and it provides an animal model of vertical transmission closely approximating human developmental timelines that could be used to investigate severe ZIKV disease outcomes and interventions in fetuses.
Collapse
Affiliation(s)
- Wilfried A A Saron
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Keerthana Shanmugam
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Chi-Ching Tung
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | | | - Abhay P S Rathore
- Department of Pathology, Duke University Medical Center, Durham, NC 27705, USA
| | - Danielle E Anderson
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
- Victorian Infectious Diseases Reference Laboratory, Melbourne, Victoria 3000, Australia
| | - Ashley L St John
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
- Department of Pathology, Duke University Medical Center, Durham, NC 27705, USA
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- SingHealth Duke-NUS Global Health Institute, Singapore 169857, Singapore
| |
Collapse
|
6
|
Gonçalves J, Melro M, Alenquer M, Araújo C, Castro-Neves J, Amaral-Silva D, Ferreira F, Ramalho JS, Charepe N, Serrano F, Pontinha C, Amorim MJ, Soares H. Balance between maternal antiviral response and placental transfer of protection in gestational SARS-CoV-2 infection. JCI Insight 2023; 8:e167140. [PMID: 37490342 PMCID: PMC10544212 DOI: 10.1172/jci.insight.167140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 07/19/2023] [Indexed: 07/27/2023] Open
Abstract
The intricate interplay between maternal immune response to SARS-CoV-2 and the transfer of protective factors to the fetus remains unclear. By analyzing mother-neonate dyads from second and third trimester SARS-CoV-2 infections, our study shows that neutralizing antibodies (NAbs) are infrequently detected in cord blood. We uncovered that this is due to impaired IgG-NAb placental transfer in symptomatic infection and to the predominance of maternal SARS-CoV-2 NAbs of the IgA and IgM isotypes, which are prevented from crossing the placenta. Crucially, the balance between maternal antiviral response and transplacental transfer of IgG-NAbs appears to hinge on IL-6 and IL-10 produced in response to SARS-CoV-2 infection. In addition, asymptomatic maternal infection was associated with expansion of anti-SARS-CoV-2 IgM and NK cell frequency. Our findings identify a protective role for IgA/IgM-NAbs in gestational SARS-CoV-2 infection and open the possibility that the maternal immune response to SARS-CoV-2 infection might benefit the neonate in 2 ways, first by skewing maternal immune response toward immediate viral clearance, and second by endowing the neonate with protective mechanisms to curtail horizontal viral transmission in the critical postnatal period, via the priming of IgA/IgM-NAbs to be transferred by the breast milk and via NK cell expansion in the neonate.
Collapse
Affiliation(s)
- Juliana Gonçalves
- Human Immunobiology and Pathogenesis Laboratory, iNOVA4Health, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| | - Magda Melro
- Human Immunobiology and Pathogenesis Laboratory, iNOVA4Health, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| | - Marta Alenquer
- Cell Biology of Viral Infection Lab, Gulbenkian Institute of Science, Oeiras, Portugal
- Católica Biomedical Research Centre, Católica Medical School, Portuguese Catholic University, Lisbon, Portugal
| | - Catarina Araújo
- Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
| | - Júlia Castro-Neves
- Human Immunobiology and Pathogenesis Laboratory, iNOVA4Health, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| | - Daniela Amaral-Silva
- Human Immunobiology and Pathogenesis Laboratory, iNOVA4Health, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| | - Filipe Ferreira
- Cell Biology of Viral Infection Lab, Gulbenkian Institute of Science, Oeiras, Portugal
- Católica Biomedical Research Centre, Católica Medical School, Portuguese Catholic University, Lisbon, Portugal
| | | | - Nádia Charepe
- Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- CHRC, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| | - Fátima Serrano
- Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- CHRC, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| | - Carlos Pontinha
- Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
| | - Maria João Amorim
- Cell Biology of Viral Infection Lab, Gulbenkian Institute of Science, Oeiras, Portugal
- Católica Biomedical Research Centre, Católica Medical School, Portuguese Catholic University, Lisbon, Portugal
| | - Helena Soares
- Human Immunobiology and Pathogenesis Laboratory, iNOVA4Health, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| |
Collapse
|
7
|
Braga C, Martelli CMT, Souza WV, Luna CF, Albuquerque MDFPM, Mariz CA, Morais CNL, Brito CAA, Melo CFCA, Lins RD, Drexler JF, Jaenisch T, Marques ETA, Viana IFT. Seroprevalence of Dengue, Chikungunya and Zika at the epicenter of the congenital microcephaly epidemic in Northeast Brazil: A population-based survey. PLoS Negl Trop Dis 2023; 17:e0011270. [PMID: 37399197 DOI: 10.1371/journal.pntd.0011270] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/03/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND The four Dengue viruses (DENV) serotypes were re-introduced in Brazil's Northeast region in a couple of decades, between 1980's and 2010's, where the DENV1 was the first detected serotype and DENV4 the latest. Zika (ZIKV) and Chikungunya (CHIKV) viruses were introduced in Recife around 2014 and led to large outbreaks in 2015 and 2016, respectively. However, the true extent of the ZIKV and CHIKV outbreaks, as well as the risk factors associated with exposure to these viruses remain vague. METHODS We conducted a stratified multistage household serosurvey among residents aged between 5 and 65 years in the city of Recife, Northeast Brazil, from August 2018 to February 2019. The city neighborhoods were stratified and divided into high, intermediate, and low socioeconomic strata (SES). Previous ZIKV, DENV and CHIKV infections were detected by IgG-based enzyme linked immunosorbent assays (ELISA). Recent ZIKV and CHIKV infections were assessed through IgG3 and IgM ELISA, respectively. Design-adjusted seroprevalence were estimated by age group, sex, and SES. The ZIKV seroprevalence was adjusted to account for the cross-reactivity with dengue. Individual and household-related risk factors were analyzed through regression models to calculate the force of infection. Odds Ratio (OR) were estimated as measure of effect. PRINCIPAL FINDINGS A total of 2,070 residents' samples were collected and analyzed. The force of viral infection for high SES were lower as compared to low and intermediate SES. DENV seroprevalence was 88.7% (CI95%:87.0-90.4), and ranged from 81.2% (CI95%:76.9-85.6) in the high SES to 90.7% (CI95%:88.3-93.2) in the low SES. The overall adjusted ZIKV seroprevalence was 34.6% (CI95%:20.0-50.9), and ranged from 47.4% (CI95%:31.8-61.5) in the low SES to 23.4% (CI95%:12.2-33.8) in the high SES. The overall CHIKV seroprevalence was 35.7% (CI95%:32.6-38.9), and ranged from 38.6% (CI95%:33.6-43.6) in the low SES to 22.3% (CI95%:15.8-28.8) in the high SES. Surprisingly, ZIKV seroprevalence rapidly increased with age in the low and intermediate SES, while exhibited only a small increase with age in high SES. CHIKV seroprevalence according to age was stable in all SES. The prevalence of serological markers of ZIKV and CHIKV recent infections were 1.5% (CI95%:0.1-3.7) and 3.5% (CI95%:2.7-4.2), respectively. CONCLUSIONS Our results confirmed continued DENV transmission and intense ZIKV and CHIKV transmission during the 2015/2016 epidemics followed by ongoing low-level transmission. The study also highlights that a significant proportion of the population is still susceptible to be infected by ZIKV and CHIKV. The reasons underlying a ceasing of the ZIKV epidemic in 2017/18 and the impact of antibody decay in susceptibility to future DENV and ZIKV infections may be related to the interplay between disease transmission mechanism and actual exposure in the different SES.
Collapse
Affiliation(s)
- Cynthia Braga
- Department of Parasitology, Institute Aggeu Magalhães, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Celina M T Martelli
- Department of Public Health, Institute Aggeu Magalhães, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Wayner V Souza
- Department of Public Health, Institute Aggeu Magalhães, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Carlos F Luna
- Department of Public Health, Institute Aggeu Magalhães, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | | | - Carolline A Mariz
- Department of Parasitology, Institute Aggeu Magalhães, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Clarice N L Morais
- Department of Virology, Institute Aggeu Magalhães, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Carlos A A Brito
- Department of Clinical Medicine, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | | | - Roberto D Lins
- Department of Virology, Institute Aggeu Magalhães, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Jan Felix Drexler
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany
- German Centre for Infection Research (DZIF), associated partner site Charité, Berlin, Germany
| | - Thomas Jaenisch
- Section Clinical Tropical Medicine, Department of Infectious Diseases, Heidelberg University Hospital, Germany
- German Centre for Infection Research (DZIF), Heidelberg Site, Heidelberg, Germany
- Center for Global Health, Colorado School of Public Health, Aurora, Colorado, United States of America
| | - Ernesto T A Marques
- Department of Virology, Institute Aggeu Magalhães, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Isabelle F T Viana
- Department of Virology, Institute Aggeu Magalhães, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| |
Collapse
|
8
|
Transplacental transfer of SARS-CoV-2 antibodies: a cohort study. Eur J Clin Microbiol Infect Dis 2023; 42:277-285. [PMID: 36692603 PMCID: PMC9872727 DOI: 10.1007/s10096-023-04553-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 01/16/2023] [Indexed: 01/25/2023]
Abstract
The purpose of this study was to examine the transfer rate of SARS-CoV-2 IgG antibodies in pregnancy and newborns. Two Danish labor wards screened all women for SARS-CoV-2 by PCR upon arrival. Women (n = 99) with a SARS-CoV-2 PCR-positive nasopharyngeal (NP) swab or with a household member with a positive swab at labor or any time during pregnancy, or COVID-19 symptoms upon admission (November 2020 through August 2021), were included. Mother and infant were tested by NP swabs at delivery, and maternal and infant (umbilical cord) venous blood samples were collected. We obtained clinical information including previous PCR test results from the medical records. SARS-Cov-2 IgM and quantified IgG antibodies were measured by enzyme-linked immunosorbent assay and transfer ratios of IgG. We detected IgG antibodies in 73 women and 65 cord blood sera and found a strong correlation between SARS-CoV-2 IgG concentrations in maternal and umbilical cord sera (r = 0.9; p < 0.05). Transfer ratio was > 1.0 in 51 out of 73 (69%) infants and > 1.5 in 26 (35%). We found that transfer was proportional to time from a positive SARS-CoV-2 PCR NP swab to delivery (r = 0.5; p < 0.05). Transfer ratios of SARS-CoV-2 antibodies were associated with time from infection to delivery with transfer ratios of more than 1.0 in the majority of seropositive mother-infant dyads.
Collapse
|
9
|
Ribeiro JF, Melo JR, Santos CDL, Chaves CR, Cabral Filho PE, Pereira G, Santos BS, Pereira GA, Rosa DS, Ribeiro RT, Fontes A. Sensitive Zika Biomarker Detection Assisted by Quantum Dot-Modified Electrochemical Immunosensing Platform. Colloids Surf B Biointerfaces 2022; 221:112984. [DOI: 10.1016/j.colsurfb.2022.112984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 09/04/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
|
10
|
Rubio R, Aguilar R, Bustamante M, Muñoz E, Vázquez-Santiago M, Santano R, Vidal M, Melero NR, Parras D, Serra P, Santamaria P, Carolis C, Izquierdo L, Gómez-Roig MD, Dobaño C, Moncunill G, Mazarico E. Maternal and neonatal immune response to SARS-CoV-2, IgG transplacental transfer and cytokine profile. Front Immunol 2022; 13:999136. [PMID: 36238312 PMCID: PMC9552073 DOI: 10.3389/fimmu.2022.999136] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/05/2022] [Indexed: 11/25/2022] Open
Abstract
SARS-CoV-2 infected pregnant women are at increased risk of severe COVID-19 than non-pregnant women and have a higher risk of adverse pregnancy outcomes like intrauterine/fetal distress and preterm birth. However, little is known about the impact of SARS-CoV-2 infection on maternal and neonatal immunological profiles. In this study, we investigated the inflammatory and humoral responses to SARS-CoV-2 in maternal and cord blood paired samples. Thirty-six pregnant women were recruited at delivery at Hospital Sant Joan de Déu, Barcelona, Spain, between April-August 2020, before having COVID-19 available vaccines. Maternal and pregnancy variables, as well as perinatal outcomes, were recorded in questionnaires. Nasopharyngeal swabs and maternal and cord blood samples were collected for SARS-CoV-2 detection by rRT-PCR and serology, respectively. We measured IgM, IgG and IgA levels to 6 SARS-CoV-2 antigens (spike [S], S1, S2, receptor-binding domain [RBD], nucleocapsid [N] full-length and C-terminus), IgG to N from 4 human coronaviruses (OC43, HKU1, 229E and NL63), and the concentrations of 30 cytokines, chemokines and growth factors by Luminex. Mothers were classified as infected or non-infected based on the rRT-PCR and serology results. Sixty-four % of pregnant women were infected with SARS-CoV-2 (positive by rRT-PCR during the third trimester and/or serology just after delivery). None of the newborns tested positive for rRT-PCR. SARS-CoV-2 infected mothers had increased levels of virus-specific antibodies and several cytokines. Those with symptoms had higher cytokine levels. IFN-α was increased in cord blood from infected mothers, and in cord blood of symptomatic mothers, EGF, FGF, IL-17 and IL-15 were increased, whereas RANTES was decreased. Maternal IgG and cytokine levels showed positive correlations with their counterparts in cord blood. rRT-PCR positive mothers showed lower transfer of SARS-CoV-2-specific IgGs, with a stronger effect when infection was closer to delivery. SARS-CoV-2 infected mothers carrying a male fetus had higher antibody levels and higher EGF, IL-15 and IL-7 concentrations. Our results show that SARS-CoV-2 infection during the third trimester of pregnancy induces a robust antibody and cytokine response at delivery and causes a significant reduction of the SARS-CoV-2-specific IgGs transplacental transfer, with a stronger negative effect when the infection is closer to delivery.
Collapse
Affiliation(s)
- Rocío Rubio
- Barcelona Institute for Global Health, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Ruth Aguilar
- Barcelona Institute for Global Health, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Mariona Bustamante
- Barcelona Institute for Global Health, Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Center for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Erica Muñoz
- Barcelona Center for Maternal-Fetal and Neonatal Medicine (BCNatal), Hospital Sant Joan de Déu and Hospital Clínic, Institut de Recerca Sant Joan de Déu (IR-SJD), Barcelona, Spain
| | - Miquel Vázquez-Santiago
- Barcelona Institute for Global Health, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Rebeca Santano
- Barcelona Institute for Global Health, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Marta Vidal
- Barcelona Institute for Global Health, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Natalia Rodrigo Melero
- Biomolecular screening and Protein Technologies Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Daniel Parras
- Pathogenesis and treatment of autoimmunity department, Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Pau Serra
- Pathogenesis and treatment of autoimmunity department, Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Pere Santamaria
- Pathogenesis and treatment of autoimmunity department, Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Julia McFarlane Diabetes Research Centre (JMDRC), and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Carlo Carolis
- Biomolecular screening and Protein Technologies Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Luis Izquierdo
- Barcelona Institute for Global Health, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Barcelona Institute for Global Health, CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Maria Dolores Gómez-Roig
- Barcelona Center for Maternal-Fetal and Neonatal Medicine (BCNatal), Hospital Sant Joan de Déu and Hospital Clínic, Institut de Recerca Sant Joan de Déu (IR-SJD), Barcelona, Spain
| | - Carlota Dobaño
- Barcelona Institute for Global Health, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Barcelona Institute for Global Health, CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
- *Correspondence: Gemma Moncunill, ; Carlota Dobaño,
| | - Gemma Moncunill
- Barcelona Institute for Global Health, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Barcelona Institute for Global Health, CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
- *Correspondence: Gemma Moncunill, ; Carlota Dobaño,
| | - Edurne Mazarico
- Barcelona Center for Maternal-Fetal and Neonatal Medicine (BCNatal), Hospital Sant Joan de Déu and Hospital Clínic, Institut de Recerca Sant Joan de Déu (IR-SJD), Barcelona, Spain
| |
Collapse
|
11
|
Lobkowicz L, Miranda-Filho DDB, Montarroyos UR, Martelli CMT, de Araújo TVB, De Souza WV, Bezerra LCA, Dhalia R, Marques ETA, Sanchez Clemente N, Webster J, Vaughan A, Webb EL, Brickley EB, Ximenes RADA. Co-circulation of Chikungunya Virus during the 2015-2017 Zika Virus Outbreak in Pernambuco, Brazil: An Analysis of the Microcephaly Epidemic Research Group Pregnancy Cohort. Am J Trop Med Hyg 2022; 106:tpmd210449. [PMID: 35405646 PMCID: PMC9209936 DOI: 10.4269/ajtmh.21-0449] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 01/20/2022] [Indexed: 12/05/2022] Open
Abstract
Co-circulation of arthropod-borne viruses, particularly those with shared mosquito vectors like Zika (ZIKV) and Chikungunya (CHIKV), is increasingly reported. An accurate differential diagnosis between ZIKV and CHIKV is of high clinical importance, especially in the context of pregnancy, but remains challenging due to limitations in the availability of specialized laboratory testing facilities. Using data collected from the prospective pregnancy cohort study of the Microcephaly Epidemic Research Group, which followed up pregnant persons with rash during the peak and decline of the 2015-2017 ZIKV epidemic in Recife, Pernambuco, Brazil, this study aims to describe the geographic and temporal distribution of ZIKV and CHIKV infections and to investigate the extent to which ZIKV and CHIKV infections may be clinically differentiable. Between December 2015 and June 2017, we observed evidence of co-circulation with laboratory confirmation of 213 ZIKV mono-infections, 55 CHIKV mono-infections, and 58 sequential ZIKV/CHIKV infections (i.e., cases with evidence of acute ZIKV infection with concomitant serological evidence of recent CHIKV infection). In logistic regressions with adjustment for maternal age, ZIKV mono-infected cases had lower odds than CHIKV mono-infected cases of presenting with arthralgia (aOR, 99% CI: 0.33, 0.15-0.74), arthritis (0.35, 0.14-0.85), fatigue (0.40, 0.17-0.96), and headache (0.44, 0.19-1.90). However, sequential ZIKV/CHIKV infections complicated discrimination, as they did not significantly differ in clinical presentation from CHIKV mono-infections. These findings suggest clinical symptoms alone may be insufficient for differentiating between ZIKV and CHIKV infections during pregnancy and therefore laboratory diagnostics continue to be a valuable tool for tailoring care in the event of arboviral co-circulation.
Collapse
Affiliation(s)
- Ludmila Lobkowicz
- Health Equity Action Lab, Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | | | | | | | | | | | - Rafael Dhalia
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Recife, Brasil
| | - Ernesto T. A. Marques
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Recife, Brasil
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nuria Sanchez Clemente
- Health Equity Action Lab, Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Jayne Webster
- Department of Disease Control, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Aisling Vaughan
- Health Equity Action Lab, Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Emily L. Webb
- MRC International Statistics and Epidemiology Group, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Elizabeth B. Brickley
- Health Equity Action Lab, Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Ricardo Arraes de Alencar Ximenes
- Departamento de Medicina Interna, Universidade de Pernambuco, Recife, Brasil
- Departamento de Medicina Tropical, Universidade Federal de Pernambuco, Recife, Brasil
| |
Collapse
|
12
|
Santiago HC, Pereira-Neto TA, Gonçalves-Pereira MH, Terzian ACB, Durbin AP. Peculiarities of Zika Immunity and Vaccine Development: Lessons from Dengue and the Contribution from Controlled Human Infection Model. Pathogens 2022; 11:pathogens11030294. [PMID: 35335618 PMCID: PMC8951202 DOI: 10.3390/pathogens11030294] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/10/2022] [Accepted: 02/15/2022] [Indexed: 01/27/2023] Open
Abstract
The Zika virus (ZIKV) was first isolated from a rhesus macaque in the Zika forest of Uganda in 1947. Isolated cases were reported until 2007, when the first major outbreaks of Zika infection were reported from the Island of Yap in Micronesia and from French Polynesia in 2013. In 2015, ZIKV started to circulate in Latin America, and in 2016, ZIKV was considered by WHO to be a Public Health Emergency of International Concern due to cases of Congenital Zika Syndrome (CZS), a ZIKV-associated complication never observed before. After a peak of cases in 2016, the infection incidence dropped dramatically but still causes concern because of the associated microcephaly cases, especially in regions where the dengue virus (DENV) is endemic and co-circulates with ZIKV. A vaccine could be an important tool to mitigate CZS in endemic countries. However, the immunological relationship between ZIKV and other flaviviruses, especially DENV, and the low numbers of ZIKV infections are potential challenges for developing and testing a vaccine against ZIKV. Here, we discuss ZIKV vaccine development with the perspective of the immunological concerns implicated by DENV-ZIKV cross-reactivity and the use of a controlled human infection model (CHIM) as a tool to accelerate vaccine development.
Collapse
Affiliation(s)
- Helton C. Santiago
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 30270-901, MG, Brazil; (T.A.P.-N.); (M.H.G.-P.)
- Correspondence: ; Tel.: +55-31-3409-2664
| | - Tertuliano A. Pereira-Neto
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 30270-901, MG, Brazil; (T.A.P.-N.); (M.H.G.-P.)
| | - Marcela H. Gonçalves-Pereira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 30270-901, MG, Brazil; (T.A.P.-N.); (M.H.G.-P.)
| | - Ana C. B. Terzian
- Laboratory of Cellular Immunology, Rene Rachou Institute, Fiocruz, Belo Horizonte 30190-002, MG, Brazil;
| | - Anna P. Durbin
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA;
| |
Collapse
|
13
|
Pratama NR, Wafa IA, Budi DS, Putra M, Wardhana MP, Wungu CDK. mRNA Covid-19 vaccines in pregnancy: A systematic review. PLoS One 2022; 17:e0261350. [PMID: 35108277 PMCID: PMC8809595 DOI: 10.1371/journal.pone.0261350] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 11/30/2021] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Pregnancy is a known risk factor for severe Coronavirus disease 2019. It is important to develop safe vaccines that elicit strong maternal and fetal antibody responses. METHODS Registries (ClinicalTrials.gov, the WHO Clinical Trial Registry, and the European Union Clinical Trial Registry) and databases (MEDLINE, ScienceDirect, Cochrane Library, Proquest, Springer, medRxiv, and bioRxiv) were systematically searched in June 20-22, 2021, for research articles pertaining to Covid-19 and pregnancy. Manual searches of bioRxiv and medRxiv were also conducted. Inclusion criteria were studies that focused on Covid-19 vaccination among pregnant women, while review articles and non-human studies were excluded. Infection rate, maternal antibody response, transplacental antibody transfer, and adverse events were described. RESULTS There were 13 observational studies with a total of 48,039 pregnant women who received mRNA vaccines. Of those, three studies investigated infection rate, six studies investigated maternal antibody response, seven studies investigated antibody transfer, three studies reported local adverse events, and five studies reported systemic adverse events. The available data suggested that the mRNA-based vaccines (Pfizer-BioNTech and Moderna) can prevent future SARS-CoV-2 infection. These vaccines did not show clear harm in pregnancy. The most commonly encountered adverse reactions were pain at the injection site, fatigue, and headache, but these were transient. Antibody responses were rapid after the first vaccine dose. After the booster, antibody responses were stronger and associated with better transplacental antibody transfer. Longer intervals between first vaccination dose and delivery were also associated with higher antibody fetal IgG and a better antibody transfer ratio. CONCLUSIONS The SARS-CoV-2 mRNA vaccines are encouraged for pregnancy. These vaccines can be a safe option for pregnant women and their fetuses. Two vaccine doses are recommended for more robust maternal and fetal antibody responses. Longer latency is associated with higher fetal antibody responses. Further research about its long-term effect on pregnancy is needed. SYSTEMATIC REVIEW REGISTRATION PROSPERO (CRD42021261684).
Collapse
Affiliation(s)
| | - Ifan Ali Wafa
- Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | | | - Manesha Putra
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America
| | - Manggala Pasca Wardhana
- Department of Obstetrics & Gynecology, Maternal-Fetal Medicine, Dr. Soetomo General Hospital, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Citrawati Dyah Kencono Wungu
- Department of Physiology and Medical Biochemistry, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
14
|
A Systematic Evaluation of IgM and IgG Antibody Assay Accuracy in Diagnosing Acute Zika Virus Infection in Brazil: Lessons Relevant to Emerging Infections. J Clin Microbiol 2021; 59:e0289320. [PMID: 34550810 DOI: 10.1128/jcm.02893-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Accurate diagnostics underpin effective public health responses to emerging viruses. For viruses, such as Zika virus (ZIKV), where the viremia clears quickly, antibody-based (IgM or IgG) diagnostics are recommended for patients who present 7 days after symptom onset. However, cross-reactive antibody responses can complicate test interpretation among populations where closely related viruses circulate. We examined the accuracy (proportion of samples correctly categorized as Zika positive or negative) for antibody-based diagnostics among Brazilian residents (Rio de Janeiro) during the ZIKV outbreak. Four ZIKV enzyme-linked immunosorbent assays (ELISAs; IgM and IgG Euroimmun, IgM Novagnost, and CDC MAC), two dengue ELISAs (IgM and IgG Panbio), and the ZIKV plaque reduction neutralization test (PRNT) were evaluated. Positive samples were ZIKV PCR confirmed clinical cases collected in 2015-2016 (n = 169); negative samples (n = 236) were collected before ZIKV was present in Brazil (≤2013). Among serum samples collected ≥7 days from symptom onset, PRNT exhibited the highest accuracy (93.7%), followed by the Euroimmun IgG ELISA (77.9%). All IgM assays exhibited lower accuracy (<75%). IgG was detected more consistently than IgM among ZIKV cases using Euroimmun ELISAs (68% versus 22%). Anti-dengue virus IgM ELISA was positive in 41.1% of confirmed ZIKV samples tested. The Euroimmun IgG assay, although misdiagnosing 22% of samples, provided the most accurate ELISA. Anti-ZIKV IgG was detected more reliably than IgM among ZIKV patients, suggesting a secondary antibody response to assay antigens following ZIKV infection. Antibody ELISAs need careful evaluation in their target population to optimize use and minimize misdiagnosis, prior to widespread deployment, particularly where related viruses cocirculate.
Collapse
|
15
|
Wilder-Smith A, Brickley EB, Ximenes RADA, Miranda-Filho DDB, Turchi Martelli CM, Solomon T, Jacobs BC, Pardo CA, Osorio L, Parra B, Lant S, Willison HJ, Leonhard S, Turtle L, Ferreira MLB, de Oliveira Franca RF, Lambrechts L, Neyts J, Kaptein S, Peeling R, Boeras D, Logan J, Dolk H, Orioli IM, Neumayr A, Lang T, Baker B, Massad E, Preet R. The legacy of ZikaPLAN: a transnational research consortium addressing Zika. Glob Health Action 2021; 14:2008139. [PMID: 35377284 PMCID: PMC8986226 DOI: 10.1080/16549716.2021.2008139] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Global health research partnerships with institutions from high-income countries and low- and middle-income countries are one of the European Commission's flagship programmes. Here, we report on the ZikaPLAN research consortium funded by the European Commission with the primary goal of addressing the urgent knowledge gaps related to the Zika epidemic and the secondary goal of building up research capacity and establishing a Latin American-European research network for emerging vector-borne diseases. Five years of collaborative research effort have led to a better understanding of the full clinical spectrum of congenital Zika syndrome in children and the neurological complications of Zika virus infections in adults and helped explore the origins and trajectory of Zika virus transmission. Individual-level data from ZikaPLAN`s cohort studies were shared for joint analyses as part of the Zika Brazilian Cohorts Consortium, the European Commission-funded Zika Cohorts Vertical Transmission Study Group, and the World Health Organization-led Zika Virus Individual Participant Data Consortium. Furthermore, the legacy of ZikaPLAN includes new tools for birth defect surveillance and a Latin American birth defect surveillance network, an enhanced Guillain-Barre Syndrome research collaboration, a de-centralized evaluation platform for diagnostic assays, a global vector control hub, and the REDe network with freely available training resources to enhance global research capacity in vector-borne diseases.
Collapse
Affiliation(s)
- Annelies Wilder-Smith
- Department of Epidemiology and Global Health, Umeå University, Umeå, Sweden.,Heidelberg Institute of Global Health, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | - Tom Solomon
- NIHR Health Protection Research Unit for Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences University of Liverpool, Liverpool, UK
| | - Bart C Jacobs
- Departments of Neurology and Immunology, Erasmus Universitair Medisch Centrum Rotterdam, The Netherlands
| | - Carlos A Pardo
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | | | | - Suzannah Lant
- NIHR Health Protection Research Unit for Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences University of Liverpool, Liverpool, UK
| | - Hugh J Willison
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK
| | - Sonja Leonhard
- Departments of Neurology and Immunology, Erasmus Universitair Medisch Centrum Rotterdam, The Netherlands
| | - Lance Turtle
- NIHR Health Protection Research Unit for Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences University of Liverpool, Liverpool, UK
| | | | | | - Louis Lambrechts
- Insect-Virus Interactions Unit, Institut Pasteur, UMR2000, CNRS, 75015 Paris, France
| | - Johan Neyts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Suzanne Kaptein
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Rosanna Peeling
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | - James Logan
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Helen Dolk
- Centre for Maternal, Fetal and Infant Research, Institute for Nursing and Health Research, Ulster University, Ulster, United Kingdom
| | - Ieda M Orioli
- RELAMC and ECLAMC at Genetics Department, Federal University of Rio de Janeiro, Brazil
| | - Andreas Neumayr
- Department of Medicine, Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Trudie Lang
- The Global Health Network, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Bonny Baker
- The Global Health Network, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Eduardo Massad
- School of Medicine, University of Sao Paulo and Fundacao Getulio Vargas, Sao Paulo, Brazil
| | - Raman Preet
- Department of Epidemiology and Global Health, Umeå University, Umeå, Sweden
| |
Collapse
|
16
|
Crooks CM, Weiler AM, Rybarczyk SL, Bliss MI, Jaeger AS, Murphy ME, Simmons HA, Mejia A, Fritsch MK, Hayes JM, Eickhoff JC, Mitzey AM, Razo E, Braun KM, Brown EA, Yamamoto K, Shepherd PM, Possell A, Weaver K, Antony KM, Morgan TK, Newman CM, Dudley DM, Schultz-Darken N, Peterson E, Katzelnick LC, Balmaseda A, Harris E, O’Connor DH, Mohr EL, Golos TG, Friedrich TC, Aliota MT. Previous exposure to dengue virus is associated with increased Zika virus burden at the maternal-fetal interface in rhesus macaques. PLoS Negl Trop Dis 2021; 15:e0009641. [PMID: 34329306 PMCID: PMC8357128 DOI: 10.1371/journal.pntd.0009641] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 08/11/2021] [Accepted: 07/09/2021] [Indexed: 11/19/2022] Open
Abstract
Concerns have arisen that pre-existing immunity to dengue virus (DENV) could enhance Zika virus (ZIKV) disease, due to the homology between ZIKV and DENV and the observation of antibody-dependent enhancement (ADE) among DENV serotypes. To date, no study has examined the impact of pre-existing DENV immunity on ZIKV pathogenesis during pregnancy in a translational non-human primate model. Here we show that macaques with a prior DENV-2 exposure had a higher burden of ZIKV vRNA in maternal-fetal interface tissues as compared to DENV-naive macaques. However, pre-existing DENV immunity had no detectable impact on ZIKV replication kinetics in maternal plasma, and all pregnancies progressed to term without adverse outcomes or gross fetal abnormalities detectable at delivery. Understanding the risks of ADE to pregnant women worldwide is critical as vaccines against DENV and ZIKV are developed and licensed and as DENV and ZIKV continue to circulate.
Collapse
Affiliation(s)
- Chelsea M. Crooks
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Andrea M. Weiler
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Sierra L. Rybarczyk
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Mason I. Bliss
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Anna S. Jaeger
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, Minnesota, United States of America
| | - Megan E. Murphy
- Department of Comparative Biosciences, 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
| | - Michael K. Fritsch
- 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
| | - Jens C. Eickhoff
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Ann M. Mitzey
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Elaina Razo
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Katarina M. Braun
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Elizabeth A. Brown
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Keisuke Yamamoto
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Phoenix M. Shepherd
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Amber Possell
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Kara Weaver
- Wisconsin National Primate Research Center, 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
| | - Terry K. Morgan
- Department of Pathology, Oregon Health and Science University, Portland, Oregon, United States of America
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Christina M. Newman
- Department of Pathology and Laboratory Medicine, 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
| | - Nancy Schultz-Darken
- 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
| | - Leah C. Katzelnick
- Division of Infectious Diseases and Vaccinology, University of California Berkeley, Berkeley, California, United States of America
| | | | - Eva Harris
- Division of Infectious Diseases and Vaccinology, University of California Berkeley, Berkeley, California, United States of America
| | - David H. O’Connor
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Emma L. Mohr
- Department of Pediatrics, 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 Comparative Biosciences, 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
| | - 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
| | - Matthew T. Aliota
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, Minnesota, United States of America
| |
Collapse
|
17
|
Hung SJ, Huang SW. Contributions of Genetic Evolution to Zika Virus Emergence. Front Microbiol 2021; 12:655065. [PMID: 34025610 PMCID: PMC8137341 DOI: 10.3389/fmicb.2021.655065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/12/2021] [Indexed: 12/15/2022] Open
Abstract
Mosquito-borne Zika virus (ZIKV) was considered an obscure virus causing only mild or self-limited symptoms until the explosive outbreaks in French Polynesia in 2013–2014 and in the Americas in 2015–2016, resulting in more than 700,000 cases of the disease, with occasional miscarriage and severe congenital birth defects, such as intrauterine growth restriction, fetal microcephaly, and other neurodevelopmental malformations. In this review, we summarized the evolution of ZIKV from a mundane virus to an epidemic virus. ZIKV has acquired a panel of amino acid substitutions during evolution when the virus spread from Africa, Asia, Pacific, through to the Americas. Robust occurrence of mutations in the evolution of ZIKV has increased its epidemic potential. Here we discussed the contributions of these evolutionary mutations to the enhancement of viral pathogenicity and host-mosquito transmission. We further explored the potential hypotheses for the increase in ZIKV activity in recent decades. Through this review, we also explored the hypotheses for the occurrence of the recent ZIKV epidemics and highlighted the potential roles of various factors including pathogen-, host-, vector-related, and environmental factors, which may have synergistically contributed to the ZIKV epidemics.
Collapse
Affiliation(s)
- Su-Jhen Hung
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Tainan, Taiwan
| | - Sheng-Wen Huang
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Tainan, Taiwan
| |
Collapse
|
18
|
Jacques IJAA, Katz L, Sena MA, Guimarães ABG, Silva YL, Albuquerque GDM, Pereira RO, de Albuquerque CAMC, Silva MAL, Oliveira PAS, Albuquerque MDFPM, Cordeiro MT, Marques ETA, França RFO, Martelli CMT, Castanha PMS, Braga C. High Incidence of Zika or Chikungunya Infection among Pregnant Women Hospitalized Due to Obstetrical Complications in Northeastern Brazil-Implications for Laboratory Screening in Arbovirus Endemic Area. Viruses 2021; 13:v13050744. [PMID: 33922819 PMCID: PMC8145990 DOI: 10.3390/v13050744] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/11/2021] [Accepted: 04/20/2021] [Indexed: 12/16/2022] Open
Abstract
The diagnostic of arbovirus-related obstetric complications in high-risk pregnancy and childbirth care is challenging, especially in endemic areas. We conducted a prospective study to track active or recent Zika (ZIKV), dengue (DENV), or chikungunya (CHIKV) virus infection among hospitalized pregnant women (PW) with obstetric complications in a hospital at the epicenter of Zika outbreak and ZIKV-related microcephaly in Brazil. Clinical data and blood samples were collected at enrollment and 10 days after the admission of study participants, between October 2018 and May 2019. Further clinical data were extracted from medical records. Samples were screened by molecular and serological tests. Out of 780 participants, 93.1% (95% CI: 91.1–94.7%) presented previous DENV exposure (IgG). ZIKV, CHIKV, and/or DENV laboratory markers of recent or active infection were detected in 130 PW, yielding a prevalence of 16.6% (95% CI: 14.2–19.5%); 9.4% (95% CI: 7.4–11.7%), 7.4% (95% CI: 5.7–9.7%), and 0.38% (95% CI: 0.1–1.2%) of CHIKV, ZIKV, and DENV infections, respectively. Most ZIKV infections were detected by molecular assays (89.6%), while CHIKV infections were detected by serology (95.9%). Our findings highlight the need for arbovirus infections screening in PW with obstetrical complications, potentially associated to these infections in endemic areas regardless of the signs or symptoms suggestive of arboviral disease.
Collapse
Affiliation(s)
- Iracema J. A. A. Jacques
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz Pernambuco, Recife CEP 50740-465, PE, Brazil; (I.J.A.A.J.); (M.A.S.); (A.B.G.G.); (Y.L.S.); (M.A.L.S.); (P.A.S.O.); (M.d.F.P.M.A.); (M.T.C.); (R.F.O.F.); (C.M.T.M.)
| | - Leila Katz
- Instituto de Medicina Integral Prof. Fernando Figueira, Recife CEP 50070-550, PE, Brazil; (L.K.); (G.D.M.A.); (R.O.P.); (C.A.M.C.d.A.)
| | - Marília A. Sena
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz Pernambuco, Recife CEP 50740-465, PE, Brazil; (I.J.A.A.J.); (M.A.S.); (A.B.G.G.); (Y.L.S.); (M.A.L.S.); (P.A.S.O.); (M.d.F.P.M.A.); (M.T.C.); (R.F.O.F.); (C.M.T.M.)
| | - Ana B. G. Guimarães
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz Pernambuco, Recife CEP 50740-465, PE, Brazil; (I.J.A.A.J.); (M.A.S.); (A.B.G.G.); (Y.L.S.); (M.A.L.S.); (P.A.S.O.); (M.d.F.P.M.A.); (M.T.C.); (R.F.O.F.); (C.M.T.M.)
| | - Yasmim L. Silva
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz Pernambuco, Recife CEP 50740-465, PE, Brazil; (I.J.A.A.J.); (M.A.S.); (A.B.G.G.); (Y.L.S.); (M.A.L.S.); (P.A.S.O.); (M.d.F.P.M.A.); (M.T.C.); (R.F.O.F.); (C.M.T.M.)
| | - Gabriela D. M. Albuquerque
- Instituto de Medicina Integral Prof. Fernando Figueira, Recife CEP 50070-550, PE, Brazil; (L.K.); (G.D.M.A.); (R.O.P.); (C.A.M.C.d.A.)
| | - Raisa O. Pereira
- Instituto de Medicina Integral Prof. Fernando Figueira, Recife CEP 50070-550, PE, Brazil; (L.K.); (G.D.M.A.); (R.O.P.); (C.A.M.C.d.A.)
| | - Camila A. M. C. de Albuquerque
- Instituto de Medicina Integral Prof. Fernando Figueira, Recife CEP 50070-550, PE, Brazil; (L.K.); (G.D.M.A.); (R.O.P.); (C.A.M.C.d.A.)
| | - Maria Almerice L. Silva
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz Pernambuco, Recife CEP 50740-465, PE, Brazil; (I.J.A.A.J.); (M.A.S.); (A.B.G.G.); (Y.L.S.); (M.A.L.S.); (P.A.S.O.); (M.d.F.P.M.A.); (M.T.C.); (R.F.O.F.); (C.M.T.M.)
| | - Paula A. S. Oliveira
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz Pernambuco, Recife CEP 50740-465, PE, Brazil; (I.J.A.A.J.); (M.A.S.); (A.B.G.G.); (Y.L.S.); (M.A.L.S.); (P.A.S.O.); (M.d.F.P.M.A.); (M.T.C.); (R.F.O.F.); (C.M.T.M.)
| | - Maria de Fátima P. M. Albuquerque
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz Pernambuco, Recife CEP 50740-465, PE, Brazil; (I.J.A.A.J.); (M.A.S.); (A.B.G.G.); (Y.L.S.); (M.A.L.S.); (P.A.S.O.); (M.d.F.P.M.A.); (M.T.C.); (R.F.O.F.); (C.M.T.M.)
| | - Marli T. Cordeiro
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz Pernambuco, Recife CEP 50740-465, PE, Brazil; (I.J.A.A.J.); (M.A.S.); (A.B.G.G.); (Y.L.S.); (M.A.L.S.); (P.A.S.O.); (M.d.F.P.M.A.); (M.T.C.); (R.F.O.F.); (C.M.T.M.)
| | - Ernesto T. A. Marques
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA 15261, USA; (E.T.A.M.J.); (P.M.S.C.)
| | - Rafael F. O. França
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz Pernambuco, Recife CEP 50740-465, PE, Brazil; (I.J.A.A.J.); (M.A.S.); (A.B.G.G.); (Y.L.S.); (M.A.L.S.); (P.A.S.O.); (M.d.F.P.M.A.); (M.T.C.); (R.F.O.F.); (C.M.T.M.)
| | - Celina M. T. Martelli
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz Pernambuco, Recife CEP 50740-465, PE, Brazil; (I.J.A.A.J.); (M.A.S.); (A.B.G.G.); (Y.L.S.); (M.A.L.S.); (P.A.S.O.); (M.d.F.P.M.A.); (M.T.C.); (R.F.O.F.); (C.M.T.M.)
| | - Priscila M. S. Castanha
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA 15261, USA; (E.T.A.M.J.); (P.M.S.C.)
- Faculdade de Ciências Médicas, Universidade de Pernambuco, Recife CEP 50100-130, PE, Brazil
| | - Cynthia Braga
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz Pernambuco, Recife CEP 50740-465, PE, Brazil; (I.J.A.A.J.); (M.A.S.); (A.B.G.G.); (Y.L.S.); (M.A.L.S.); (P.A.S.O.); (M.d.F.P.M.A.); (M.T.C.); (R.F.O.F.); (C.M.T.M.)
- Instituto de Medicina Integral Prof. Fernando Figueira, Recife CEP 50070-550, PE, Brazil; (L.K.); (G.D.M.A.); (R.O.P.); (C.A.M.C.d.A.)
- Correspondence: ; Tel.: +55-81-2101-2577
| |
Collapse
|
19
|
Coutinho CM, Fernandes JDCP, Yamamoto AY, Negrini SFBDM, Negrini BVDM, Teixeira SR, Amaral FR, da Motta MSF, Bárbaro AAT, Aragon DC, Montoya M, Harris E, Duarte G, Mussi-Pinhata MM. Persistence of Anti-ZIKV-IgG over Time Is Not a Useful Congenital Infection Marker in Infants Born to ZIKV-Infected Mothers: The NATZIG Cohort. Viruses 2021; 13:711. [PMID: 33924066 PMCID: PMC8074187 DOI: 10.3390/v13040711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/05/2021] [Accepted: 04/16/2021] [Indexed: 11/23/2022] Open
Abstract
Confirming ZIKV congenital infection is challenging because viral RNA is infrequently detected. We compared the presence of anti-ZIKV-IgM and the persistence of anti-ZIKV-IgG antibodies over 18 months in two cohorts of infants born to ZIKV-infected mothers: Cohort one: 30 infants with typical microcephaly or major brain abnormalities (Congenital Zika Syndrome-CZS); Cohort two: 123 asymptomatic infants. Serum samples obtained within 6 months of age were tested for anti-ZIKV-IgM. Anti-ZIKV-IgG was quantified in sequential samples collected at birth, 3-6 weeks, 3, 6, 12, and 18 months. ZIKV-RNA was never detected postnatally. Anti-ZIKV-IgM antibodies were detected at least once in 15/25 (60.0%; 95%CI: 38.7-78.9) infants with CZS and in 2/115 (1.7%; 95%CI: 0.2-6.1) asymptomatic infants. Although anti-ZIKV-IgG was always positive within 3-6 weeks of age, IgG levels decreased similarly over time in both cohorts. IgG levels decreased similarly in ZIKV-IgM-positive and ZIKV-IgM-negative CZS infants. Differently from other congenital infections, IgM would fail to diagnose 40% of severely symptomatic infants, and the persistence of IgG is not a useful marker for discriminating congenital infection among infants exposed to maternal ZIKV infection.
Collapse
Affiliation(s)
- Conrado Milani Coutinho
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil;
| | - Juliana Dias Crivelenti Pereira Fernandes
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil; (J.D.C.P.F.); (A.Y.Y.); (S.F.B.d.M.N.); (B.V.d.M.N.); (F.R.A.); (M.S.F.d.M.); (A.A.T.B.); (D.C.A.)
| | - Aparecida Yulie Yamamoto
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil; (J.D.C.P.F.); (A.Y.Y.); (S.F.B.d.M.N.); (B.V.d.M.N.); (F.R.A.); (M.S.F.d.M.); (A.A.T.B.); (D.C.A.)
| | - Silvia Fabiana Biason de Moura Negrini
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil; (J.D.C.P.F.); (A.Y.Y.); (S.F.B.d.M.N.); (B.V.d.M.N.); (F.R.A.); (M.S.F.d.M.); (A.A.T.B.); (D.C.A.)
| | - Bento Vidal de Moura Negrini
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil; (J.D.C.P.F.); (A.Y.Y.); (S.F.B.d.M.N.); (B.V.d.M.N.); (F.R.A.); (M.S.F.d.M.); (A.A.T.B.); (D.C.A.)
| | - Sara Reis Teixeira
- Department of Imaging, Hematology and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil;
| | - Fabiana Rezende Amaral
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil; (J.D.C.P.F.); (A.Y.Y.); (S.F.B.d.M.N.); (B.V.d.M.N.); (F.R.A.); (M.S.F.d.M.); (A.A.T.B.); (D.C.A.)
| | - Márcia Soares Freitas da Motta
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil; (J.D.C.P.F.); (A.Y.Y.); (S.F.B.d.M.N.); (B.V.d.M.N.); (F.R.A.); (M.S.F.d.M.); (A.A.T.B.); (D.C.A.)
| | - Adriana Aparecida Tiraboschi Bárbaro
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil; (J.D.C.P.F.); (A.Y.Y.); (S.F.B.d.M.N.); (B.V.d.M.N.); (F.R.A.); (M.S.F.d.M.); (A.A.T.B.); (D.C.A.)
| | - Davi Casale Aragon
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil; (J.D.C.P.F.); (A.Y.Y.); (S.F.B.d.M.N.); (B.V.d.M.N.); (F.R.A.); (M.S.F.d.M.); (A.A.T.B.); (D.C.A.)
| | - Magelda Montoya
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720-7360, USA; (M.M.); (E.H.)
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720-7360, USA; (M.M.); (E.H.)
| | - Geraldo Duarte
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil;
| | - Marisa Márcia Mussi-Pinhata
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil; (J.D.C.P.F.); (A.Y.Y.); (S.F.B.d.M.N.); (B.V.d.M.N.); (F.R.A.); (M.S.F.d.M.); (A.A.T.B.); (D.C.A.)
| | | |
Collapse
|
20
|
Atyeo C, Pullen KM, Bordt EA, Fischinger S, Burke J, Michell A, Slein MD, Loos C, Shook LL, Boatin AA, Yockey LJ, Pepin D, Meinsohn MC, Nguyen NMP, Chauvin M, Roberts D, Goldfarb IT, Matute JD, James KE, Yonker LM, Bebell LM, Kaimal AJ, Gray KJ, Lauffenburger D, Edlow AG, Alter G. Compromised SARS-CoV-2-specific placental antibody transfer. Cell 2021; 184:628-642.e10. [PMID: 33476549 PMCID: PMC7755577 DOI: 10.1016/j.cell.2020.12.027] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/16/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023]
Abstract
SARS-CoV-2 infection causes more severe disease in pregnant women compared to age-matched non-pregnant women. Whether maternal infection causes changes in the transfer of immunity to infants remains unclear. Maternal infections have previously been associated with compromised placental antibody transfer, but the mechanism underlying this compromised transfer is not established. Here, we used systems serology to characterize the Fc profile of influenza-, pertussis-, and SARS-CoV-2-specific antibodies transferred across the placenta. Influenza- and pertussis-specific antibodies were actively transferred. However, SARS-CoV-2-specific antibody transfer was significantly reduced compared to influenza- and pertussis-specific antibodies, and cord titers and functional activity were lower than in maternal plasma. This effect was only observed in third-trimester infection. SARS-CoV-2-specific transfer was linked to altered SARS-CoV-2-antibody glycosylation profiles and was partially rescued by infection-induced increases in IgG and increased FCGR3A placental expression. These results point to unexpected compensatory mechanisms to boost immunity in neonates, providing insights for maternal vaccine design.
Collapse
Affiliation(s)
- Caroline Atyeo
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; PhD Program in Virology, Division of Medical Sciences, Harvard University, Boston, MA 02115, USA
| | - Krista M Pullen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Evan A Bordt
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Stephanie Fischinger
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; PhD Program in Immunology and Virology, University of Duisburg-Essen, Essen 47057, Germany
| | - John Burke
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Ashlin Michell
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Matthew D Slein
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Carolin Loos
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lydia L Shook
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Adeline A Boatin
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Laura J Yockey
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - David Pepin
- Pediatric Surgical Research Laboratories, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Marie-Charlotte Meinsohn
- Pediatric Surgical Research Laboratories, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ngoc Minh Phuong Nguyen
- Pediatric Surgical Research Laboratories, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Maeva Chauvin
- Pediatric Surgical Research Laboratories, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Drucilla Roberts
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ilona T Goldfarb
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Juan D Matute
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Kaitlyn E James
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lael M Yonker
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lisa M Bebell
- Division of Infectious Diseases, Massachusetts General Hospital, MGH Global Health, and Harvard Medical School, Boston, MA 02114, USA
| | - Anjali J Kaimal
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Kathryn J Gray
- Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Douglas Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Andrea G Edlow
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA.
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
21
|
Adam A, Cuellar S, Wang T. Memory B cell and antibody responses to flavivirus infection and vaccination. Fac Rev 2021; 10:5. [PMID: 33659923 PMCID: PMC7894259 DOI: 10.12703/r/10-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Flaviviruses are a group of mosquito- or tick-borne single-stranded RNA viruses that can cause a wide range of clinical manifestations in humans and animals, including asymptomatic, flu-like febrile illness, hemorrhagic fever, encephalitis, birth defects, and death. Many of them have no licensed vaccines available for human use. Memory B cell development and induction of neutralizing antibody responses, which are important for the control of flavivirus infection and dissemination, have been used as biomarkers for vaccine efficacy. In this review, we will discuss recent findings on memory B cells and antibody responses from studies in clinical specimen and animal models of flavivirus infection and vaccination with a focus on several clinically important flaviviruses, including dengue, West Nile, yellow fever, and Zika viruses.
Collapse
Affiliation(s)
- Awadalkareem Adam
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Servando Cuellar
- School of Medicine, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Tian Wang
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, 77555, USA
| |
Collapse
|
22
|
Ngwe Tun MM, Moriuchi M, Toizumi M, Luvai E, Raini S, Kitamura N, Takegata M, Nguyen HAT, Moi ML, Buerano CC, Anh DD, Yoshida LM, Morita K, Moriuchi H. Congenital Zika Virus Infection in a Birth Cohort in Vietnam, 2017-2018. Am J Trop Med Hyg 2021; 103:2059-2064. [PMID: 32815502 PMCID: PMC7646788 DOI: 10.4269/ajtmh.20-0286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To detect congenital ZIKV infection (CZI) in a birth cohort and among high-risk neonates in Vietnam, we collected umbilical cord blood plasma samples of newly delivered babies and peripheral plasma samples of high-risk neonates in Nha Trang, central Vietnam, between July 2017 and September 2018. Samples were subjected to serological and molecular tests. Of the 2013 newly delivered babies, 21 (1%) were positive for Zika virus (ZIKV) IgM and 1,599 (79%) for Flavivirus IgG. Among the 21 ZIKV IgM-positives, 11 were confirmed to have CZI because their plasma samples had anti-ZIKV neutralization titers ≥ 4 times higher than those against dengue virus (DENV)-1 to 4 and Japanese encephalitis virus (JEV) and were tested for the ZIKV RNA positive by real-time reverse transcription–PCR. Therefore, the incidence of CZI in our birth cohort was approximately 0.5%. Of the 150 high-risk neonates, three (2%) and 95 (63%) were positive for ZIKV IgM and Flavivirus IgG antibodies, respectively. None of the three ZIKV IgM-positives had ≥ 4 times higher anti-ZIKV neutralization titers than those against DENV-1 to 4 and JEV, and were therefore considered as probable CZI. Our results indicate that CZI is not rare in Vietnam. Although those with confirmed CZI did not show apparent symptoms suspected of congenital Zika syndrome at birth, detailed examinations and follow-up studies are needed to clarify the CZI impact in Vietnam. This is the first report of CZI cases in a birth cohort in Asia.
Collapse
Affiliation(s)
- Mya Myat Ngwe Tun
- Department of Virology, Institute of Tropical Medicine and Leading Program, Nagasaki University, Nagasaki, Japan
| | - Masako Moriuchi
- Department of Pediatrics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Michiko Toizumi
- Department of Pediatric Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Elizabeth Luvai
- Department of Virology, Institute of Tropical Medicine and Leading Program, Nagasaki University, Nagasaki, Japan
| | - Sandra Raini
- Department of Virology, Institute of Tropical Medicine and Leading Program, Nagasaki University, Nagasaki, Japan
| | - Noriko Kitamura
- Department of Pediatric Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Mizuki Takegata
- Department of Pediatric Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | | | - Meng Ling Moi
- Department of Virology, Institute of Tropical Medicine and Leading Program, Nagasaki University, Nagasaki, Japan
| | - Corazon C Buerano
- Research and Biotechnology, St. Luke's Medical Center, Quezon City, Philippines
| | - Dang Duc Anh
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - Lay-Myint Yoshida
- Department of Pediatric Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Kouichi Morita
- Department of Virology, Institute of Tropical Medicine and Leading Program, Nagasaki University, Nagasaki, Japan
| | - Hiroyuki Moriuchi
- Department of Pediatrics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
23
|
Congenital abnormalities associated with Zika virus infection-Dengue as potential co-factor? A systematic review. PLoS Negl Trop Dis 2021; 15:e0008984. [PMID: 33395432 PMCID: PMC7808571 DOI: 10.1371/journal.pntd.0008984] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 01/14/2021] [Accepted: 11/12/2020] [Indexed: 11/21/2022] Open
Abstract
Zika virus (ZIKV) emerged in Brazil during 2013–2014 causing an epidemic of previously unknown congenital abnormalities. The frequency of severe congenital abnormalities after maternal ZIKV infection revealed an unexplained geographic variability, especially between the Northeast and the rest of Brazil. Several reasons for this variability have been discussed. Prior immunity against Dengue virus (DENV) affecting ZIKV seems to be the most likely explanation. Here we summarise the current evidence regarding this prominent co-factor to potentially explain the geographic variability. This systematic review followed the PRISMA guidelines. The search was conducted up to May 15th, 2020, focussing on immunological interactions from Zika virus with previous Dengue virus infections as potential teratogenic effect for the foetus. Eight out of 339 screened studies reported on the association between ZIKV, prior DENV infection and microcephaly, mostly focusing on antibody-dependent enhancement (ADE) as potential pathomechanism. Prior DENV infection was associated with enhancement for ZIKV infection and increased neurovirulence in one included in vitro study only. Interestingly, the seven in vivo studies exhibited a heterogeneous picture with three studies showing a protective effect of prior DENV infections and others no effect at all. According to several studies, socio-economic factors are associated with increased risk for microcephaly. Very few studies addressed the question of unexplained variability of infection-related microcephaly. Many studies focussed on ADE as mechanism without measuring microcephaly as endpoint. Interestingly, three of the included studies reported a protective effect of prior DENV infection against microcephaly. This systematic review strengthens the hypothesis that immune priming after recent DENV infection is the crucial factor for determining protection or enhancement activity. It is of high importance that the currently ongoing prospective studies include a harmonised assessment of the potential candidate co-factors. Despite the large volume of Zika virus (ZIKV)-related research, the difference in the frequency of ZIKV-related severe congenital abnormalities between Northeastern Brazil in the rest of Brazil and other locations has not been adequately explained. This systematic review summarizes the existing evidence for the role of prior Dengue virus (DENV) infections as co-factor to explain the variability in the frequency of ZIKV-related severe congenital abnormalities. Results differed significantly according to study type (e.g. in vitro versus in vivo studies), and no clear picture emerged with regard to prior DENV infections causing ZIKV-related severe congenital abnormalities. However, evidence from additional studies not included in this systematic review that focus on immunological interactions between DENV and ZIKV infections imply the potential that severe ZIKV disease is more frequent in those with prior DENV infections. Therefore, an assessment of background immunity against closely related viruses (e.g. the flavivirus family) needs to be included in future and ongoing longitudinal studies on ZIKV-related congenital abnormalities.
Collapse
|
24
|
Edlow AG, Li JZ, Collier ARY, Atyeo C, James KE, Boatin AA, Gray KJ, Bordt EA, Shook LL, Yonker LM, Fasano A, Diouf K, Croul N, Devane S, Yockey LJ, Lima R, Shui J, Matute JD, Lerou PH, Akinwunmi BO, Schmidt A, Feldman J, Hauser BM, Caradonna TM, De la Flor D, D’Avino P, Regan J, Corry H, Coxen K, Fajnzylber J, Pepin D, Seaman MS, Barouch DH, Walker BD, Yu XG, Kaimal AJ, Roberts DJ, Alter G. Assessment of Maternal and Neonatal SARS-CoV-2 Viral Load, Transplacental Antibody Transfer, and Placental Pathology in Pregnancies During the COVID-19 Pandemic. JAMA Netw Open 2020; 3:e2030455. [PMID: 33351086 PMCID: PMC7756241 DOI: 10.1001/jamanetworkopen.2020.30455] [Citation(s) in RCA: 294] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 10/28/2020] [Indexed: 12/15/2022] Open
Abstract
Importance Biological data are lacking with respect to risk of vertical transmission and mechanisms of fetoplacental protection in maternal severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Objective To quantify SARS-CoV-2 viral load in maternal and neonatal biofluids, transplacental passage of anti-SARS-CoV-2 antibody, and incidence of fetoplacental infection. Design, Setting, and Participants This cohort study was conducted among pregnant women presenting for care at 3 tertiary care centers in Boston, Massachusetts. Women with reverse transcription-polymerase chain reaction (RT-PCR) results positive for SARS-CoV-2 were recruited from April 2 to June 13, 2020, and follow-up occurred through July 10, 2020. Contemporaneous participants without SARS-CoV-2 infection were enrolled as a convenience sample from pregnant women with RT-PCR results negative for SARS-CoV-2. Exposures SARS-CoV-2 infection in pregnancy, defined by nasopharyngeal swab RT-PCR. Main Outcomes and Measures The main outcomes were SARS-CoV-2 viral load in maternal plasma or respiratory fluids and umbilical cord plasma, quantification of anti-SARS-CoV-2 antibodies in maternal and cord plasma, and presence of SARS-CoV-2 RNA in the placenta. Results Among 127 pregnant women enrolled, 64 with RT-PCR results positive for SARS-CoV-2 (mean [SD] age, 31.6 [5.6] years) and 63 with RT-PCR results negative for SARS-CoV-2 (mean [SD] age, 33.9 [5.4] years) provided samples for analysis. Of women with SARS-CoV-2 infection, 23 (36%) were asymptomatic, 22 (34%) had mild disease, 7 (11%) had moderate disease, 10 (16%) had severe disease, and 2 (3%) had critical disease. In viral load analyses among 107 women, there was no detectable viremia in maternal or cord blood and no evidence of vertical transmission. Among 77 neonates tested in whom SARS-CoV-2 antibodies were quantified in cord blood, 1 had detectable immunoglobuilin M to nucleocapsid. Among 88 placentas tested, SARS-CoV-2 RNA was not detected in any. In antibody analyses among 37 women with SARS-CoV-2 infection, anti-receptor binding domain immunoglobin G was detected in 24 women (65%) and anti-nucleocapsid was detected in 26 women (70%). Mother-to-neonate transfer of anti-SARS-CoV-2 antibodies was significantly lower than transfer of anti-influenza hemagglutinin A antibodies (mean [SD] cord-to-maternal ratio: anti-receptor binding domain immunoglobin G, 0.72 [0.57]; anti-nucleocapsid, 0.74 [0.44]; anti-influenza, 1.44 [0.80]; P < .001). Nonoverlapping placental expression of SARS-CoV-2 receptors angiotensin-converting enzyme 2 and transmembrane serine protease 2 was noted. Conclusions and Relevance In this cohort study, there was no evidence of placental infection or definitive vertical transmission of SARS-CoV-2. Transplacental transfer of anti-SARS-CoV-2 antibodies was inefficient. Lack of viremia and reduced coexpression and colocalization of placental angiotensin-converting enzyme 2 and transmembrane serine protease 2 may serve as protective mechanisms against vertical transmission.
Collapse
Affiliation(s)
- Andrea G. Edlow
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston
| | - Jonathan Z. Li
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ai-ris Y. Collier
- Department of Obstetrics, Gynecology and Reproductive Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Caroline Atyeo
- Ragon Institute of MGH, MIT and Harvard, Harvard Medical School, Cambridge, Massachusetts
| | - Kaitlyn E. James
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Adeline A. Boatin
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Kathryn J. Gray
- Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Evan A. Bordt
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Lydia L. Shook
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Lael M. Yonker
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Alessio Fasano
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Khady Diouf
- Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Natalie Croul
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Samantha Devane
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Laura J. Yockey
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Rosiane Lima
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Jessica Shui
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Juan D. Matute
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Paul H. Lerou
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Babatunde O. Akinwunmi
- Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Aaron Schmidt
- Ragon Institute of MGH, MIT and Harvard, Harvard Medical School, Cambridge, Massachusetts
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts
| | - Jared Feldman
- Ragon Institute of MGH, MIT and Harvard, Harvard Medical School, Cambridge, Massachusetts
| | - Blake M. Hauser
- Ragon Institute of MGH, MIT and Harvard, Harvard Medical School, Cambridge, Massachusetts
| | - Timothy M. Caradonna
- Ragon Institute of MGH, MIT and Harvard, Harvard Medical School, Cambridge, Massachusetts
| | - Denis De la Flor
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Paolo D’Avino
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston
| | - James Regan
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Heather Corry
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kendyll Coxen
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jesse Fajnzylber
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - David Pepin
- Pediatric Surgical Research Laboratories, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Michael S. Seaman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Ragon Institute of MGH, MIT and Harvard, Harvard Medical School, Cambridge, Massachusetts
| | - Bruce D. Walker
- Ragon Institute of MGH, MIT and Harvard, Harvard Medical School, Cambridge, Massachusetts
| | - Xu G. Yu
- Ragon Institute of MGH, MIT and Harvard, Harvard Medical School, Cambridge, Massachusetts
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Anjali J. Kaimal
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Drucilla J. Roberts
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Harvard Medical School, Cambridge, Massachusetts
| |
Collapse
|
25
|
Plaque Reduction Neutralization Test (PRNT) in the Congenital Zika Syndrome: Positivity and Associations with Laboratory, Clinical, and Imaging Characteristics. Viruses 2020; 12:v12111244. [PMID: 33142747 PMCID: PMC7692785 DOI: 10.3390/v12111244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/16/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023] Open
Abstract
The short duration of viremia, low blood viral load, inaccessibility to timely specific diagnostic tests, and cross-reactions with other flaviviruses have hindered laboratory confirmation of Congenital Zika Syndrome (CZS). This study analyzes the positivity of the plaque reduction neutralization test (PRNT) in children with clinical or imaging characteristics of CZS and its association with laboratory, clinical, and imaging characteristics. The 94 clinical cases of CZS submitted to the ZIKV PRNT90 test were followed from 2016 to 2018. The mean age of children at PRNT90 collection was 22 ± 6 months Standard Deviation. The ZIKV PRNT90 was positive (titer ≥ 10) in 40 (42.5%) children. ZIKV PRNT90 positivity was associated with severe microcephaly in newborns (p = 0.016), lower head circumference z-score at birth (p = 0.043) and 24 months of age (p = 0.031), and severe reduction of the cerebral parenchyma volume (p = 0.021), expressing greater disease severity. Negative PRNT90 in children with characteristic signs of CZS may be due to false-negative results, indicating that the diagnosis of CZS should be primarily syndromic.
Collapse
|
26
|
Katzelnick LC, Bos S, Harris E. Protective and enhancing interactions among dengue viruses 1-4 and Zika virus. Curr Opin Virol 2020; 43:59-70. [PMID: 32979816 DOI: 10.1016/j.coviro.2020.08.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 07/31/2020] [Accepted: 08/10/2020] [Indexed: 12/18/2022]
Abstract
Dengue viruses 1-4 (DENV 1-4) and Zika virus (ZIKV) are closely related flaviviruses transmitted by Aedes mosquitoes that co-circulate in Asia, the Americas, Africa, and Oceania. Here, we review recent and historical literature on in vitro experiments, animal models, and clinical and epidemiological studies to describe how the sequence of DENV 1-4 and ZIKV infections modulates subsequent dengue and Zika disease outcome. Overall, we find these interactions are asymmetric. Immunity from a prior DENV infection or a prior ZIKV infection can enhance future severe dengue disease for some DENV serotypes while protecting against other serotypes. Further, prior DENV immunity has not been shown to enhance future uncomplicated or severe Zika and instead appears to be protective. Interestingly, secondary ZIKV infection induces type-specific ZIKV immunity but only generates weakly cross-neutralizing anti-DENV/ZIKV immunity, consistent with risk of future dengue disease. In contrast, secondary DENV infection induces strongly cross-neutralizing antibodies that protect against subsequent severe dengue disease. These immunologic interactions may be explained by differences in virion structure between DENV 1-4 and ZIKV, which modulate thermostability, susceptibility to neutralization, and cell infectivity. Overall, these observations are important for the understanding and prediction of epidemics and the development and evaluation of dengue and Zika vaccines.
Collapse
Affiliation(s)
- Leah C Katzelnick
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, 185 Li Ka Shing Center, 1951 Oxford Street, Berkeley, CA 94720-3370, United States.
| | - Sandra Bos
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, 185 Li Ka Shing Center, 1951 Oxford Street, Berkeley, CA 94720-3370, United States
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, 185 Li Ka Shing Center, 1951 Oxford Street, Berkeley, CA 94720-3370, United States.
| |
Collapse
|
27
|
Collier ARY, Borducchi EN, Chandrashekar A, Moseley E, Peter L, Teodoro NS, Nkolola J, Abbink P, Barouch DH. Sustained maternal antibody and cellular immune responses in pregnant women infected with Zika virus and mother to infant transfer of Zika-specific antibodies. Am J Reprod Immunol 2020; 84:e13288. [PMID: 32557984 DOI: 10.1111/aji.13288] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 06/07/2020] [Accepted: 06/09/2020] [Indexed: 01/02/2023] Open
Abstract
PROBLEM Evaluation of Zika virus (ZIKV)-specific humoral and cellular immune response in pregnant women exposed to ZIKV. METHOD OF STUDY In this observational, prospective cohort study, we recruited pregnant women presenting for prenatal ultrasound for ZIKV exposure at a single academic teaching hospital in Boston, MA from November 2016 to December 2018. We collected blood, urine, and cervicovaginal swabs antepartum, intrapartum, and postpartum; and cord blood and placenta at delivery. We used experimental assays to calculate quantitative viral loads, ZIKV-specific immunoglobulin titers, and ZIKV-specific T-cell responses. RESULTS We enrolled 22 participants, three of which had serologic-confirmed ZIKV infection. No participants demonstrated sustained ZIKV shedding. ZIKV-specific IgG/IgM antibody was sustained throughout pregnancy and postpartum. ZIKV envelope and capsid-specific T-cell responses were also observed, albeit inconsistent. No newborns in this cohort had congenital Zika syndrome. Infant cord blood of infected mothers exhibited ZIKV-specific IgG, but not IgM antibodies. CONCLUSION We detected a robust, prolonged maternal humoral immune response to ZIKV during pregnancy and postpartum. We also demonstrated evidence for efficient transplacental antibody transfer from mother to infant at birth, supporting the importance of neonatal passive immunity to ZIKV. Maternal T-cell responses were less consistent among pregnant women infected with ZIKV.
Collapse
Affiliation(s)
- Ai-Ris Y Collier
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Erica N Borducchi
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Abishek Chandrashekar
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Edward Moseley
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Lauren Peter
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Nicholas S Teodoro
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Joseph Nkolola
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Peter Abbink
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
28
|
Putri ND, Dhenni R, Handryastuti S, Johar E, Ma’roef CN, Fadhilah A, Perma Iskandar AT, Prayitno A, Karyanti MR, Satari HI, Jumiyanti N, Aprilia YY, Sriyani IY, Dewi YP, Yudhaputri FA, Safari D, Hadinegoro SR, Rosenberg R, Powers AM, Aye Myint KS. Absence of Evidence of Zika Virus Infection in Cord Blood and Urine from Newborns with Congenital Abnormalities, Indonesia. Am J Trop Med Hyg 2020; 102:876-879. [PMID: 32043460 PMCID: PMC7124925 DOI: 10.4269/ajtmh.19-0593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 11/20/2019] [Indexed: 11/07/2022] Open
Abstract
Zika virus (ZIKV) has recently been confirmed as endemic in Indonesia, but no congenital anomalies (CA) related to ZIKV infection have been reported. We performed molecular and serological testing for ZIKV and other flaviviruses on cord serum and urine samples collected in October 2016 to April 2017 during a prospective, cross-sectional study of neonates in Jakarta, Indonesia. Of a total of 429 neonates, 53 had CA, including 14 with microcephaly. These 53, and 113 neonate controls without evidence of CA, were tested by ZIKV-specific real-time reverse transcription polymerase chain reaction (RT-PCR), pan-flavivirus RT-PCR, anti-ZIKV and anti-DENV IgM ELISA, and plaque reduction neutralization test. There was no evidence of ZIKV infection among neonates in either the CA or non-CA cohorts, except in three cases with low titers of anti-ZIKV neutralizing antibodies. Further routine evaluation throughout Indonesia of pregnant women and their newborns for exposure to ZIKV should be a high priority for determining risk.
Collapse
Affiliation(s)
- Nina Dwi Putri
- Department of Paediatrics, Dr. Cipto Mangunkusumo National Central Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Rama Dhenni
- Emerging Virus Research Unit, Eijkman Institute for Molecular Biology, Jakarta, Indonesia
| | - Setyo Handryastuti
- Department of Paediatrics, Dr. Cipto Mangunkusumo National Central Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Edison Johar
- Emerging Virus Research Unit, Eijkman Institute for Molecular Biology, Jakarta, Indonesia
| | - Chairin Nisa Ma’roef
- Emerging Virus Research Unit, Eijkman Institute for Molecular Biology, Jakarta, Indonesia
| | - Araniy Fadhilah
- Emerging Virus Research Unit, Eijkman Institute for Molecular Biology, Jakarta, Indonesia
| | - Adhi Teguh Perma Iskandar
- Department of Paediatrics, Dr. Cipto Mangunkusumo National Central Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Ari Prayitno
- Department of Paediatrics, Dr. Cipto Mangunkusumo National Central Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Mulya Rahma Karyanti
- Department of Paediatrics, Dr. Cipto Mangunkusumo National Central Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Hindra Irawan Satari
- Department of Paediatrics, Dr. Cipto Mangunkusumo National Central Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Niphidiah Jumiyanti
- Department of Paediatrics, Dr. Cipto Mangunkusumo National Central Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Yuni Yudha Aprilia
- Department of Paediatrics, Dr. Cipto Mangunkusumo National Central Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Ida Yus Sriyani
- Emerging Virus Research Unit, Eijkman Institute for Molecular Biology, Jakarta, Indonesia
| | - Yora Permata Dewi
- Emerging Virus Research Unit, Eijkman Institute for Molecular Biology, Jakarta, Indonesia
| | | | - Dodi Safari
- Emerging Virus Research Unit, Eijkman Institute for Molecular Biology, Jakarta, Indonesia
| | - Sri Rezeki Hadinegoro
- Department of Paediatrics, Dr. Cipto Mangunkusumo National Central Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Ronald Rosenberg
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado
| | - Ann M. Powers
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado
| | - Khin Saw Aye Myint
- Emerging Virus Research Unit, Eijkman Institute for Molecular Biology, Jakarta, Indonesia
| |
Collapse
|
29
|
Hurtado-Monzón AM, Cordero-Rivera CD, Farfan-Morales CN, Osuna-Ramos JF, De Jesús-González LA, Reyes-Ruiz JM, Del Ángel RM. The role of anti-flavivirus humoral immune response in protection and pathogenesis. Rev Med Virol 2020; 30:e2100. [PMID: 32101633 DOI: 10.1002/rmv.2100] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 12/15/2022]
Abstract
Flavivirus infections are a public health threat in the world that requires the development of safe and effective vaccines. Therefore, the understanding of the anti-flavivirus humoral immune response is fundamental to future studies on flavivirus pathogenesis and the design of anti-flavivirus therapeutics. This review aims to provide an overview of the current understanding of the function and involvement of flavivirus proteins in the humoral immune response as well as the ability of the anti-envelope (anti-E) antibodies to interfere (neutralizing antibodies) or not (non-neutralizing antibodies) with viral infection, and how they can, in some circumstances enhance dengue virus infection on Fc gamma receptor (FcγR) bearing cells through a mechanism known as antibody-dependent enhancement (ADE). Thus, the dual role of the antibodies against E protein poses a formidable challenge for vaccine development. Also, we discuss the roles of antibody binding stoichiometry (the concentration, affinity, or epitope recognition) in the neutralization of flaviviruses and the "breathing" of flavivirus virions in the humoral immune response. Finally, the relevance of some specific antibodies in the design and improvement of effective vaccines is addressed.
Collapse
Affiliation(s)
- Arianna Mahely Hurtado-Monzón
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - Carlos Daniel Cordero-Rivera
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - Carlos Noe Farfan-Morales
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - Juan Fidel Osuna-Ramos
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - Luis Adrián De Jesús-González
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - José Manuel Reyes-Ruiz
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - Rosa María Del Ángel
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| |
Collapse
|
30
|
Carvalho MDCG, Ximenes RAA, Montarroyos UR, da Silva PFS, Andrade-Valença LPA, Eickmann SH, Ramos RC, Rocha MÂW, de Araujo TVB, de Albuquerque MDFPM, Martelli CMT, de Souza WV, Brickley EB, Miranda-Filho DDB. Early epilepsy in children with Zika-related microcephaly in a cohort in Recife, Brazil: Characteristics, electroencephalographic findings, and treatment response. Epilepsia 2020; 61:509-518. [PMID: 32065676 PMCID: PMC7155083 DOI: 10.1111/epi.16444] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 01/22/2020] [Accepted: 01/22/2020] [Indexed: 01/08/2023]
Abstract
Objective To estimate the incidence of epilepsy in children with Zika‐related microcephaly in the first 24 months of life; to characterize the associated clinical and electrographic findings; and to summarize the treatment responses. Methods We followed a cohort of children, born during the 2015‐2016 Zika virus (ZIKV) epidemic in Brazil, with congenital microcephaly and evidence of congenital ZIKV infection on neuroimaging and/or laboratory testing. Neurological assessments were performed at ≤3, 6, 12, 15, 18, 21, and 24 months of life. Serial electroencephalograms were performed over the first 24 months. Results We evaluated 91 children, of whom 48 were female. In this study sample, the cumulative incidence of epilepsy was 71.4% in the first 24 months, and the main type of seizure was infantile spasms (83.1%). The highest incidence of seizures occurred between 3 and 9 months of age, and the risk remained high until 15 months of age. The incidence of infantile spasms peaked between 4 and 7 months and was followed by an increased incidence of focal epilepsy cases after 12 months of age. Neuroimaging results were available for all children, and 100% were abnormal. Cortical abnormalities were identified in 78.4% of the 74 children evaluated by computed tomography and 100% of the 53 children evaluated by magnetic resonance imaging. Overall, only 46.1% of the 65 children with epilepsy responded to treatment. The most commonly used medication was sodium valproate with or without benzodiazepines, levetiracetam, phenobarbital, and vigabatrin. Significance Zika‐related microcephaly was associated with high risk of early epilepsy. Seizures typically began after the third month of life, usually as infantile spasms, with atypical electroencephalographic abnormalities. The seizure control rate was low. The onset of seizures in the second year was less frequent and, when it occurred, presented as focal epilepsy.
Collapse
Affiliation(s)
| | - Ricardo A A Ximenes
- University of Pernambuco, Recife, Brazil.,Federal University of Pernambuco, Recife, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Cerebrospinal fluid immunoglobulins are increased in neonates exposed to Zika virus during foetal life. J Infect 2020; 80:419-425. [PMID: 31981639 DOI: 10.1016/j.jinf.2020.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/18/2019] [Accepted: 01/14/2020] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To compare immunoglobulin levels in cerebrospinal fluid (CSF) of neonates exposed to Zika virus (ZIKV) during foetal life (cases) with levels in CSF of control neonates. METHODS We identified 16 neonates who underwent lumbar puncture (LP), during the ZIKV epidemic (December/2015 to March/2016) whose mothers reported ZIKV clinical symptoms during gestation (cases). Congenital microcephaly was defined as head circumference ≤31.9 cm (boys) and ≤31.5 cm (girls) for term neonates, or ≤2 standard deviations below the mean for premature (<37 weeks) neonates. Subsequently, we identified neonates who underwent LP in the same lab and fulfilled criteria to be controls: age ≤4 days, CSF white blood cell count ≤8/mm3, CSF protein ≤132 mg/dL, CSF red blood cell count ≤1,000/mm3, neither central nervous system illness, nor congenital infection, nor microcephaly. CSF immunoglobulin concentrations were measured by mass spectrometry. RESULTS 13 controls were included. IgM, IgA, IgG, IgK, and IgL were significantly higher among cases (p < 0.001). Eight (50%) ZIKV exposed infants had congenital microcephaly. These showed the strongest immunoglobulin elevation of the IgM and IgA classes. CONCLUSION Neonates exposed to ZIKV infection during gestation present with elevated distinct immunoglobulins in CSF, both in cases that developed microcephaly and in cases that did not.
Collapse
|
32
|
Ximenes RADA, Miranda-Filho DDB, Brickley EB, Montarroyos UR, Martelli CMT, de Araújo TVB, Rodrigues LC, de Albuquerque MDFPM, de Souza WV, Castanha PMDS, França RFO, Dhália R, Marques ETA. Zika virus infection in pregnancy: Establishing a case definition for clinical research on pregnant women with rash in an active transmission setting. PLoS Negl Trop Dis 2019; 13:e0007763. [PMID: 31589611 PMCID: PMC6797234 DOI: 10.1371/journal.pntd.0007763] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 10/17/2019] [Accepted: 09/08/2019] [Indexed: 12/26/2022] Open
Abstract
Defining cases of Zika virus (ZIKV) infection is a critical challenge for epidemiological research. Due to ZIKV's overlapping clinical features and potential immunologic cross-reactivity with other flaviviruses and the current lack of an optimal ZIKV-specific diagnostic assay, varying approaches for identifying ZIKV infections have been employed to date. This paper presents the laboratory results and diagnostic criteria developed by the Microcephaly Epidemic Research Group for defining cases of maternal ZIKV infection in a cohort of pregnant women with rash (N = 694) recruited during the declining 2015-2017 epidemic in northeast Brazil. For this investigation, we tested maternal sera for ZIKV by quantitative reverse transcription polymerase chain reaction (qRT-PCR), Immunoglobulin (Ig) M and IgG3 enzyme-linked immunosorbent assays (ELISAs), and Plaque Reduction Neutralization Test (PRNT50). Overall, 23.8% of participants tested positive by qRT-PCR during pregnancy (range of detection: 0-72 days after rash onset). However, the inter-assay concordance was lower than expected. Among women with qRT-PCR-confirmed ZIKV and further testing, only 10.1% had positive IgM tests within 90 days of rash, and only 48.5% had ZIKV-specific PRNT50 titers ≥20 within 1 year of rash. Given the complexity of these data, we convened a panel of experts to propose an algorithm for identifying ZIKV infections in pregnancy based on all available lines of evidence. When the diagnostic algorithm was applied to the cohort, 26.9% of participants were classified as having robust evidence of a ZIKV infection during pregnancy, 4.0% as having moderate evidence, 13.3% as having limited evidence of a ZIKV infection but with uncertain timing, and 19.5% as having evidence of an unspecified flavivirus infection before or during pregnancy. Our findings suggest that integrating longitudinal data from nucleic acid and serologic testing may enhance diagnostic sensitivity and underscore the need for an on-going dialogue regarding the optimization of strategies for defining cases of ZIKV in research.
Collapse
Affiliation(s)
- Ricardo Arraes de Alencar Ximenes
- Departamento de Medicina Tropical, Universidade Federal de Pernambuco, Recife, PE, Brasil
- Departamento de Medicina Interna, Universidade de Pernambuco, Recife, PE, Brasil
| | | | - Elizabeth B. Brickley
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | | | | | - Laura C. Rodrigues
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | | | - Priscila Mayrelle da Silva Castanha
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Recife, PE, Brasil
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Faculdade de Ciências Médicas, Universidade de Pernambuco, Recife, PE, Brasil
| | | | - Rafael Dhália
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Recife, PE, Brasil
| | - Ernesto T. A. Marques
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Recife, PE, Brasil
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | | |
Collapse
|
33
|
|
34
|
Efficient transplacental IgG transfer in women infected with Zika virus during pregnancy. PLoS Negl Trop Dis 2019; 13:e0007648. [PMID: 31449521 PMCID: PMC6730934 DOI: 10.1371/journal.pntd.0007648] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 09/06/2019] [Accepted: 07/22/2019] [Indexed: 12/28/2022] Open
Abstract
Zika virus (ZIKV) is a newly-identified infectious cause of congenital disease. Transplacental transfer of maternal IgG to the fetus plays an important role in preventing many neonatal infections. However, antibody transfer may also have negative consequences, such as mediating enhancement of flavivirus infections in early life, or trafficking of virus immune complexes to the fetal compartment. ZIKV infection produces placental pathology which could lead to impaired IgG transfer efficiency as occurs in other maternal infections, such as HIV-1 and malaria. In this study, we asked whether ZIKV infection during pregnancy impairs transplacental transfer of IgG. We enrolled pregnant women with fever or rash in a prospective cohort in Vitoria, Brazil during the recent ZIKV epidemic. ZIKV and dengue virus (DENV)-specific IgG, ZIKV and DENV neutralizing antibodies, and routine vaccine antigen-specific IgG were measured in maternal samples collected around delivery and 20 paired cord blood samples. We concluded that 8 of these mothers were infected with ZIKV during pregnancy and 12 were ZIKV-uninfected. The magnitude of flavivirus-specific IgG, neutralizing antibody, and vaccine-elicited IgG were highly correlated between maternal plasma and infant cord blood in both ZIKV-infected and -uninfected mother-infant pairs. Moreover, there was no difference in the magnitude of plasma flavivirus-specific IgG levels between mothers and infants regardless of ZIKV infection status. Our data suggests that maternal ZIKV infection during pregnancy does not impair the efficiency of placental transfer of flavivirus-specific, functional, and vaccine-elicited IgG. These findings have implications for the neonatal outomes of maternal ZIKV infection and optimal administration of antibody-based ZIKV vaccines and therapeutics.
Collapse
|