Immune and pathophysiologic profiling of antenatal coronavirus disease 2019 in the GIFT cohort: A Singaporean case-control study

Yue Gu, Jia Ming Low, Jolene Su Yi Tan, Melissa Shu Feng Ng, Lisa F P Ng, Bhuvaneshwari Shunmuganathan, Rashi Gupta, Paul A MacAry, Zubair Amin, Le Ye Lee, Derrick Lian, Lynette Pei-Chi Shek, Youjia Zhong, Liang Wei Wang, Yue Gu, Jia Ming Low, Jolene Su Yi Tan, Melissa Shu Feng Ng, Lisa F P Ng, Bhuvaneshwari Shunmuganathan, Rashi Gupta, Paul A MacAry, Zubair Amin, Le Ye Lee, Derrick Lian, Lynette Pei-Chi Shek, Youjia Zhong, Liang Wei Wang

Abstract

COVID-19 can be severe in pregnant women, and have adverse consequences for the subsequent infant. We profiled the post-infectious immune responses in maternal and child blood as well as breast milk in terms of antibody and cytokine expression and performed histopathological studies on placentae obtained from mothers convalescent from antenatal COVID-19. Seventeen mother-child dyads (8 cases of antenatal COVID-19 and 9 healthy unrelated controls; 34 individuals in total) were recruited to the Gestational Immunity For Transfer (GIFT) study. Maternal and infant blood, and breast milk samples were collected over the first year of life. All samples were analyzed for IgG and IgA against whole SARS-CoV-2 spike protein, the spike receptor-binding domain (RBD), and previously reported immunodominant epitopes, as well as cytokine levels. The placentae were examined microscopically. The study is registered at clinicaltrials.gov under the identifier NCT04802278. We found high levels of virus-specific IgG in convalescent mothers and similarly elevated titers in newborn children. Thus, antenatal SARS-CoV-2 infection led to high plasma titers of virus-specific antibodies in infants postnatally. However, this waned within 3-6 months of life. Virus neutralization by plasma was not uniformly achieved, and the presence of antibodies targeting known immunodominant epitopes did not assure neutralization. Virus-specific IgA levels were variable among convalescent individuals' sera and breast milk. Antibody transfer ratios and the decay of transplacentally transferred virus-specific antibodies in neonatal circulation resembled that for other pathogens. Convalescent mothers showed signs of chronic inflammation marked by persistently elevated IL17RA levels in their blood. Four placentae presented signs of acute inflammation, particularly in the subchorionic region, marked by neutrophil infiltration even though > 50 days had elapsed between virus clearance and delivery. Administration of a single dose of BNT162b2 mRNA vaccine to mothers convalescent from antenatal COVID-19 increased virus-specific IgG and IgA titers in breast milk, highlighting the importance of receiving the vaccine even after natural infection with the added benefit of enhanced passive immunity.

Keywords: GIFT; SARS-CoV-2; antenatal COVID-19; breast milk antibodies; placental inflammation; transplacental antibody transfer.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Gu, Low, Tan, Ng, Ng, Shunmuganathan, Gupta, MacAry, Amin, Lee, Lian, Shek, Zhong and Wang.

Figures

FIGURE 1
FIGURE 1
Neutrophilic infiltration present in the placentae of convalescent mothers. Microscopic analysis shows a neutrophilic infiltrate in the subchorionic region of the placentae of three patients—(A) patient CS2, H&E × 100, (B) patient CS3, H&E ×100, (C) patient CS7, H&E × 100. (D) Higher power view of the inflammatory process (patient CS3, H&E × 200).
FIGURE 2
FIGURE 2
Spike and RBD-specific IgG and IgA present in maternal plasma of convalescent mothers. Using a protein-based ELISA, maternal plasma from control (CT01- CT10) and convalescent (CS01-CS07) mothers obtained 1-month post-partum were used for the detection of spike (A,C) and RBD (B,D) specific IgG and IgA antibodies. The bar graphs represent the normalized average signals of antibodies binding to the respective proteins. Convalescent mothers (CS samples) are arranged in ascending order according to their time from COVID diagnosis to delivery.
FIGURE 3
FIGURE 3
Neutralization of SARS-CoV-2 pseudovirus using maternal plasma. Plasma from control (n = 10) and convalescent mothers (n = 7). Bar graphs represent the average percentage neutralization. Convalescent mothers (CS samples) are arranged in ascending order according to their time from COVID diagnosis to delivery.
FIGURE 4
FIGURE 4
Spike and RBD-specific IgG are passively transferred from convalescent mothers to infants. (A) Spike and (B) RBD-specific IgG from maternal plasma 1 month post-partum and at three time points from babies’ plasma was determined using ELISA. Transfer ratio at the 1 month timepoint of (C) spike and (D) RBD-specific IgG from mother to infant was calculated by taking the averaged IgG signal of baby’s over the mother’s IgG. Convalescent mothers (CS samples) are arranged in ascending order according to their time from COVID diagnosis to delivery.
FIGURE 5
FIGURE 5
Spike and RBD-specific IgA present in maternal breast milk of convalescent mothers. Using a protein-based ELISA, maternal breast milk from control (CT01- CT10) and convalescent (CS01-CS08) mothers obtained at two time points 0–1 months and 2–3 months post-partum were used for the detection of spike and RBD-specific IgG and IgA antibodies. Breast milk was screened for (A) spike-specific IgG, (B) RBD-specific IgG, (C) spike-specific IgA, and (D) RBD-specific IgA. The bar graphs represent the normalized average signals of antibodies binding to the respective proteins. Convalescent mothers (CS samples) are arranged in ascending order according to their time from COVID diagnosis to delivery.
FIGURE 6
FIGURE 6
Cytokine profile differences in healthy and convalescent mothers’ plasma. Using 1-month post-partum plasma (n = 6 for controls and n = 7 for cases), cytokines were evaluated using the 41-plex Luminex™ assay. (A) A representation of all cytokines that were detected, (B) IL-17RA levels and (C) MCP-1. Convalescent mothers are represented in red while healthy controls are represented in black. Bar graphs represent the average log(concentration) from the samples from the respective groups in A while the measured concentrations were used for (B,C). *p < 0.05.
FIGURE 7
FIGURE 7
Increased secretion and production of spike-specific IgA and IgG antibodies post-vaccination in convalescent mothers. Breast milk from three nursing convalescent mothers were collected at 4 time points, prior to vaccination (0), 3 days post-vaccination, 7 days post-vaccination, and 30 days post-vaccination for a (A,C) spike and (B,D) RBD protein-based IgA and IgG ELISA assay. The x-axis refers to the number of days post-vaccination.

References

    1. Eastin C, Eastin T. Epidemiological characteristics of 2143 pediatric patients with 2019 coronavirus disease in China: Dong Y, Mo X, Hu Y et al. Pediatrics. 2020; J Emerg Med. (2020) 58:712–3. 10.1542/peds.2020-0702
    1. Kyle MH, Glassman ME, Khan A, Fernández CR, Hanft E, Emeruwa UN, et al. A review of newborn outcomes during the COVID-19 pandemic. Semin Perinatol. (2020) 44:151286. 10.1016/j.semperi.2020.151286
    1. Cacho NT, Lawrence RM. Innate immunity and breast milk. Front Immunol. (2017) 8:584. 10.3389/fimmu.2017.00584
    1. Demers-Mathieu V, Underwood MA, Beverly RL, Nielsen SD, Dallas DC. Comparison of human milk immunoglobulin survival during gastric digestion between preterm and term infants. Nutrients. (2018) 10:631. 10.3390/nu10050631
    1. Iyer AS, Jones FK, Nodoushani A, Kelly M, Becker M, Slater D, et al. Dynamics and significance of the antibody response to SARS-CoV-2 infection. medRxiv [Preprint]. (2020). 10.1101/2020.07.18.20155374
    1. Zhao J, Yuan Q, Wang H, Liu W, Liao X, Su Y, et al. Antibody responses to SARS-CoV-2 in patients with novel coronavirus disease 2019. Clin Infect Dis. (2020) 71:2027–34. 10.1093/cid/ciaa344
    1. Dong Y, Chi X, Hai H, Sun L, Zhang M, Xie WF, et al. Antibodies in the breast milk of a maternal woman with COVID-19. Emerg Microbes Infect. (2020) 9:1467–9. 10.1080/22221751.2020.1780952
    1. Fox A, Marino J, Amanat F, Krammer F, Hahn-Holbrook J, Zolla-Pazner S, et al. Robust and specific secretory IgA against SARS-CoV-2 detected in human milk. iScience. (2020) 23:101735. 10.1016/j.isci.2020.101735
    1. Gao X, Wang S, Zeng W, Chen S, Wu J, Lin X, et al. Clinical and immunologic features among COVID-19-affected mother-infant pairs: antibodies to SARS-CoV-2 detected in breast milk. New Microbes New Infect. (2020) 37:100752. 10.1016/j.nmni.2020.100752
    1. Raghuvamsi PV, Tulsian NK, Samsudin F, Qian X, Purushotorman K, Yue G, et al. SARS-CoV-2 S protein:ACE2 interaction reveals novel allosteric targets. Elife. (2021) 10:e63646. 10.7554/eLife.63646
    1. Kongsuphol P, Jia H, Cheng HL, Gu Y, Shunmuganathan BDO, Chen MW, et al. A rapid simple point-of-care assay for the detection of SARS-CoV-2 neutralizing antibodies. Commun Med. (2021) 1:46. 10.1038/s43856-021-00045-9
    1. Amrun SN, Lee CY, Lee B, Fong SW, Young BE, Chee RS, et al. Linear B-cell epitopes in the spike and nucleocapsid proteins as markers of SARS-CoV-2 exposure and disease severity. EBioMedicine. (2020) 58:102911. 10.1016/j.ebiom.2020.102911
    1. Poh CM, Carissimo G, Wang B, Amrun SN, Lee CY-P, Chee RS-L, et al. Two linear epitopes on the SARS-CoV-2 spike protein that elicit neutralising antibodies in COVID-19 patients. Nat Commun. (2020) 11:2806. 10.1038/s41467-020-16638-2
    1. Chan YH, Young BE, Fong SW, Ding Y, Goh YS, Chee RS, et al. Differential cytokine responses in hospitalized COVID-19 patients limit efficacy of remdesivir. Front Immunol. (2021) 12:680188. 10.3389/fimmu.2021.680188
    1. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. (2020) 395:497–506. 10.1016/S0140-6736(20)30183-5
    1. Chan JF, Yuan S, Kok KH, To KK, Chu H, Yang J, et al. A familial cluster of pneumonia associated with the 2019 novel coronavirus indicating person-to-person transmission: a study of a family cluster. Lancet. (2020) 395:514–23. 10.1016/S0140-6736(20)30154-9
    1. Fan C, Guo Y, Qu P, Wang S, Wang M, Yuan J, et al. No obviously adverse pregnancy complications and outcomes of the recovered pregnant women from COVID-19. Reprod Toxicol. (2021) 100:163–6. 10.1016/j.reprotox.2020.11.008
    1. Sharps MC, Hayes DJL, Lee S, Zou Z, Brady CA, Almoghrabi Y, et al. A structured review of placental morphology and histopathological lesions associated with SARS-CoV-2 infection. Placenta. (2020) 101:13–29. 10.1016/j.placenta.2020.08.018
    1. Di Girolamo R, Khalil A, Alameddine S, D’Angelo E, Galliani C, Matarrelli B, et al. Placental histopathology after SARS-CoV-2 infection in pregnancy: a systematic review and meta-analysis. Am J Obstetr Gynecol. (2021) 3:100468. 10.1016/j.ajogmf.2021.100468
    1. Shrock E, Fujimura E, Kula T, Timms RT, Lee IH, Leng Y, et al. Viral epitope profiling of COVID-19 patients reveals cross-reactivity and correlates of severity. Science. (2020) 370:eabd4250.
    1. Flannery DD, Gouma S, Dhudasia MB, Mukhopadhyay S, Pfeifer MR, Woodford EC, et al. Assessment of maternal and neonatal cord blood SARS-CoV-2 antibodies and placental transfer ratios. JAMA Pediatr. (2021) 175:594–600. 10.1001/jamapediatrics.2021.0038
    1. Atyeo C, Pullen KM, Bordt EA, Fischinger S, Burke J, Michell A, et al. Compromised SARS-CoV-2-specific placental antibody transfer. Cell. (2021) 184:628–42.e10. 10.1016/j.cell.2020.12.027
    1. Fu C, Lu L, Wu H, Shaman J, Cao Y, Fang F, et al. Placental antibody transfer efficiency and maternal levels: specific for measles, coxsackievirus A16, enterovirus 71, poliomyelitis I-III and HIV-1 antibodies. Sci Rep. (2016) 6:38874. 10.1038/srep38874
    1. Zervou FN, Louie P, Stachel A, Zacharioudakis IM, Ortiz-Mendez Y, Thomas K, et al. SARS-CoV-2 antibodies: IgA correlates with severity of disease in early COVID-19 infection. J Med Virol. (2021) 93:5409–15. 10.1002/jmv.27058
    1. Ma H, Zeng W, He H, Zhao D, Jiang D, Zhou P, et al. Serum IgA, IgM, and IgG responses in COVID-19. Cell Mol Immunol. (2020) 17:773–5. 10.1038/s41423-020-0474-z
    1. Schwartz DA, Morotti D. Placental pathology of COVID-19 with and without fetal and neonatal infection: trophoblast necrosis and chronic histiocytic intervillositis as risk factors for transplacental transmission of SARS-CoV-2. Viruses. (2020) 12:1308. 10.3390/v12111308
    1. Menter T, Mertz KD, Jiang S, Chen H, Monod C, Tzankov A, et al. Placental pathology findings during and after SARS-CoV-2 Infection: features of villitis and malperfusion. Pathobiology. (2021) 88:69–77. 10.1159/000511324
    1. Facchetti F, Bugatti M, Drera E, Tripodo C, Sartori E, Cancila V, et al. SARS-CoV2 vertical transmission with adverse effects on the newborn revealed through integrated immunohistochemical, electron microscopy and molecular analyses of placenta. EBioMedicine. (2020) 59:102951. 10.1016/j.ebiom.2020.102951
    1. Palmeira P, Quinello C, Silveira-Lessa AL, Zago CA, Carneiro-Sampaio M. IgG placental transfer in healthy and pathological pregnancies. Clin Dev Immunol. (2012) 2012:985646. 10.1155/2012/985646
    1. Xu Z, Shi L, Wang Y, Zhang J, Huang L, Zhang C, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med. (2020) 8:420–2. 10.1016/S2213-2600(20)30076-X
    1. Pacha O, Sallman MA, Evans SE. COVID-19: a case for inhibiting IL-17? Nat Rev Immunol. (2020) 20:345–6. 10.1038/s41577-020-0328-z
    1. Ong SWX, Fong SW, Young BE, Chan YH, Lee B, Amrun SN, et al. Persistent symptoms and association with inflammatory cytokine signatures in recovered coronavirus disease 2019 patients. Open Forum Infect Dis. (2021) 8:ofab156. 10.1093/ofid/ofab156
    1. Cacciapuoti S, De Rosa A, Gelzo M, Megna M, Raia M, Pinchera B, et al. Immunocytometric analysis of COVID patients: a contribution to personalized therapy? Life Sci. (2020) 261:118355. 10.1016/j.lfs.2020.118355
    1. Deshmane SL, Kremlev S, Amini S, Sawaya BE. Monocyte chemoattractant protein-1 (MCP-1): an overview. J Interferon Cytokine Res. (2009) 29:313–26. 10.1089/jir.2008.0027
    1. Xi X, Guo Y, Zhu M, Wei Y, Li G, Du B, et al. Higher expression of monocyte chemotactic protein 1 in mild COVID-19 patients might be correlated with inhibition of type I IFN signaling. Virol J. (2021) 18:12. 10.1186/s12985-020-01478-9
    1. Chen X, Pan Z, Yue S, Yu F, Zhang J, Yang Y, et al. Disease severity dictates SARS-CoV-2-specific neutralizing antibody responses in COVID-19. Signal Transduct Target Ther. (2020) 5:180. 10.1038/s41392-020-00301-9
    1. Cai Y, Zhang J, Xiao T, Peng H, Sterling SM, Walsh RM, Jr, et al. Distinct conformational states of SARS-CoV-2 spike protein. Science. (2020) 369:1586–92. 10.1126/science.abd4251

Source: PubMed

3
구독하다