1
|
Balla J, Rathore APS, St. John AL. Maternal IgE Influence on Fetal and Infant Health. Immunol Rev 2025; 331:e70029. [PMID: 40281548 PMCID: PMC12032057 DOI: 10.1111/imr.70029] [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: 03/24/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025]
Abstract
Immunoglobulin E (IgE) is the most recently discovered and evolved mammalian antibody type, best known for interacting with mast cells (MCs) as immune effectors. IgE-mediated antigen sensing by MC provides protection from parasites, venomous animals, bacteria, and other insults to barrier tissues exposed to the environment. IgE and MCs act as inflammation amplifiers and immune response adjuvants. Thus, IgE production and memory formation are greatly constrained and require specific licensing. Failure of regulation gives rise to allergic disease, one of the top causes of chronic illness. Increasing evidence suggests allergy development often starts early in life, including prenatally, with maternal influence being central in shaping the offspring's immune system. Although IgE often exists before birth, an endogenous source of IgE-producing B cells has not been identified. This review discusses the mechanisms of maternal IgE transfer into the offspring, its interactions with offspring MCs and antigen-presenting cells, and the consequences for allergic inflammation and allergen sensitization development. We discuss the multifaceted effects of pre-existing IgG, IgE, and their glycosylation on maternal IgE transfer and functionality in the progeny. Understanding the IgE-mediated mechanisms predisposing for early life allergy development may allow their targeting with existing therapeutics and guide the development of new ones.
Collapse
Affiliation(s)
- Jozef Balla
- Programme in Emerging Infectious DiseasesDuke‐National University of Singapore Medical SchoolSingaporeSingapore
| | - Abhay P. S. Rathore
- Programme in Emerging Infectious DiseasesDuke‐National University of Singapore Medical SchoolSingaporeSingapore
- Department of PathologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Ashley L. St. John
- Programme in Emerging Infectious DiseasesDuke‐National University of Singapore Medical SchoolSingaporeSingapore
- Department of PathologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- SingHealth Duke‐NUS Global Health InstituteSingaporeSingapore
| |
Collapse
|
2
|
Bentes AA, Mara Gonçalves de Oliveira Azevedo V, Lemos SMA, Pinheiro GSMA, de Araújo Martins I, Cotta NHS, Martins dos Santos Oliveira R, Lousado Mesquita G, Januário GC, Januário JN, Shankar AH, Lindgren Alves CR. Factors modulating maternofetal transfer of IgG antibodies following SARS-CoV-2 gestational infection. Rev Inst Med Trop Sao Paulo 2025; 67:e29. [PMID: 40243801 PMCID: PMC11996034 DOI: 10.1590/s1678-9946202567029] [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: 11/18/2024] [Accepted: 03/06/2025] [Indexed: 04/18/2025] Open
Abstract
Early infant immunity to SARS-CoV-2 depends on maternofetal transfer of antibodies. We aimed to analyze the factors modulating the maternofetal transfer of anti-SARS-CoV-2 IgG antibodies following gestational infection during the pandemic in Brazil (April-August 2021). We conducted a retrospective and prospective cohort study involving 509 mother-child dyads tested simultaneously for IgG anti-nucleocapsid antibodies during universal neonatal screening. There were 341 seronegative dyads and 168 seropositive ones. Seropositive neonates were retested two to three months later. We examined the association of neonatal serological status and IgG concentrations with gestational mRNA vaccination, timing of maternal infection, neonatal conditions, and gender. Gestational SARS-CoV-2 infection predicted neonatal IgG seropositivity (OR=3.97; 95%CI=2.69-5.88). Maternal infection in the first, second, or third trimester was associated with progressively greater seropositivity in neonates (34.4%, 51.6%, and 58.2%, respectively; p=0.03). Among seropositive neonates, IgG concentration was higher when mothers reported they had COVID-19 during pregnancy (p=0.04) and tended to be lower in girls (p=0.06). More than half of the seropositive neonates remained seropositive two to three months later (54.1%), which was associated with both maternal and neonatal IgG concentration at birth (p<0.001). Higher neonatal IgG concentrations at birth were associated with the persistence of anti-N IgG antibodies for two to three months in more than half of the seropositive newborns. This study provides an additional understanding of the dynamics of maternofetal antibody transfer.
Collapse
Affiliation(s)
- Aline Almeida Bentes
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Departamento de Pediatria, Belo Horizonte, Minas Gerais, Brazil
| | | | - Stela Maris Aguiar Lemos
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Departamento de Fonoaudiologia, Belo Horizonte, Minas Gerais, Brazil
| | - Gabriela Soutto Mayor Assumpção Pinheiro
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Programa de Pós-Graduação em Ciências da Saúde - Saúde da Criança e do Adolescente, Belo Horizonte, Minas Gerais, Brazil
| | - Isadora de Araújo Martins
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Programa de Pós-Graduação em Ciências da Saúde - Saúde da Criança e do Adolescente, Belo Horizonte, Minas Gerais, Brazil
| | | | | | - Gabriela Lousado Mesquita
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Belo Horizonte, Minas Gerais, Brazil
- Secretaria de Saúde do Estado de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gabriela Cintra Januário
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Departamento de Clínica Médica, Núcleo de Ações e Pesquisa em Apoio Diagnóstico, Belo Horizonte, Minas Gerais, Brazil
| | - José Nélio Januário
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Departamento de Pediatria, Belo Horizonte, Minas Gerais, Brazil
| | - Anuraj H. Shankar
- University of Oxford, Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, Oxford, United Kingdom
- Oxford University, Clinical Research Unit, Jakarta, Indonesia
| | - Claudia Regina Lindgren Alves
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Departamento de Pediatria, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
3
|
Sturrock S, Cavell B, Alexander F, Apostolakis K, Barro C, Daniel O, Dixon L, Halkerston R, Hall T, Hesp JR, Hill AM, Leung S, Lim S, McStraw N, Otter A, Ramkhelawon L, Watts R, Etti M, Heath P, Lee-Wo C, Greening V, Khalil A, Turner K, Taylor S, Doare KL, Ladhani S. Maternal and Placental Antibody Responses in SARS-CoV-2 Vaccination and Natural Infection During Pregnancy. Pediatr Infect Dis J 2025; 44:S32-S37. [PMID: 39951071 PMCID: PMC7617455 DOI: 10.1097/inf.0000000000004704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
BACKGROUND As COVID-19 becomes endemic, understanding antibody response and transfer during pregnancy is crucial to inform policy and vaccination schedules. While good immunogenicity has been shown from SARS-CoV-2 vaccines, few data are available demonstrating functional responses in pregnant populations and infants. METHODS A prospective, multi-site observational study was completed across 14 centers in England from April 23, 2020, to December 21, 2022. Demographic, COVID infection and vaccination data were collected. Maternal and cord blood samples were taken at delivery, with maternal and neonatal blood samples taken at 6 weeks for participants who had been infected or vaccinated. Antibody concentrations were measured using antibody-dependent complement deposition, antibody-dependent neutrophil phagocytosis, ACE2 inhibition and Roche and EuroImmun antibody binding assays at the UK Health Security Agency. RESULTS Maternal vaccination and infection both produced an antibody response in 100% of mothers and 93.8% and 92.9% of neonates, respectively, which persisted at 6 weeks in 95%. The strongest response was seen in mothers who were both vaccinated and infected. Anti-spike antibody response decreased almost 25-fold from first to third trimester vaccination (P=0.013). Placental transfer of antibodies post-infection showed varied results depending on the assay used, with higher transfer ratios observed in assays measuring Fc-mediated antibody effector functions and IgG-specific responses. CONCLUSIONS Maternal vaccination is associated with good immunogenicity and successful antibody transfer to the neonate, particularly with vaccination in early pregnancy. Further study is needed to determine the mechanism by which the timing of vaccination affects antibody transfer. When measuring placental transfer of antibodies, consideration of the assay to use is essential.
Collapse
Affiliation(s)
- Sarah Sturrock
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| | | | | | | | - Camille Barro
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| | - Olwenn Daniel
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| | | | | | - Tom Hall
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| | | | | | | | - Suzy Lim
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| | | | | | - Laxmee Ramkhelawon
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| | - Robert Watts
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| | - Melanie Etti
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| | - Paul Heath
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| | - Chelone Lee-Wo
- St George’s Hospital NHS Trust, UK
- St George’s, University of London, UK
| | - Vanessa Greening
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| | | | - Kim Turner
- Keck School of Medicine, University of Southern California
| | | | - Kirsty Le Doare
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
- Makerere University-Johns Hopkins University Research Collaboration, Uganda
| | - Shamez Ladhani
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| |
Collapse
|
4
|
Hu J, Huynh DT, Boyce M. Sugar Highs: Recent Notable Breakthroughs in Glycobiology. Biochemistry 2024; 63:2937-2947. [PMID: 39475524 DOI: 10.1021/acs.biochem.4c00418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
Glycosylation is biochemically complex and functionally critical to a wide range of processes and disease states, making it a vibrant area of contemporary research. Here, we highlight a selection of notable recent advances in the glycobiology of SARS-CoV-2 infection and immunity, cancer biology and immunotherapy, and newly discovered glycosylated RNAs. Together, these studies illustrate the significance of glycosylation in normal biology and the great promise of manipulating glycosylation for therapeutic benefit in disease.
Collapse
Affiliation(s)
- Jimin Hu
- Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina 27710, United States
| | - Duc T Huynh
- Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina 27710, United States
| | - Michael Boyce
- Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina 27710, United States
| |
Collapse
|
5
|
Jasset OJ, Lopez Zapana PA, Bahadir Z, Shook L, Dennis M, Gilbert E, Liu ZA, Yinger RV, Bald C, Bradford CG, Silfen AH, Klein SL, Pekosz A, Permar S, Konnikova L, Yonker LM, Lauffenburger D, Nelson A, Elovitz MA, Edlow AG. Enhanced placental antibody transfer efficiency with longer interval between maternal respiratory syncytial virus vaccination and birth. Am J Obstet Gynecol 2024:S0002-9378(24)01125-6. [PMID: 39515450 PMCID: PMC12056162 DOI: 10.1016/j.ajog.2024.10.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/26/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Respiratory syncytial virus is associated with significant neonatal and infant morbidity and mortality. Maternal bivalent respiratory syncytial virus prefusion F respiratory syncytial virus vaccination to protect neonates and infants was approved in September 2023 for administration between 32+0 and 36+6 weeks to protect neonates and infants. This approved timeframe is narrower than the 24 to 36 week window evaluated in the clinical trial, due to the possible association between preterm birth and vaccine administration. Currently, data are lacking on how maternal vaccine timing within the approved window affects the transfer of antibodies from mother to fetus, critical information that could influence clinical practice. OBJECTIVE We sought to examine how gestational age at vaccination and time elapsed from maternal respiratory syncytial virus vaccination to delivery impacted transfer of maternal antibodies measured in the umbilical cord at delivery and in peripheral blood of 2-month infants. We also examined differences in maternal and cord respiratory syncytial virus antibody levels achieved by vaccination vs natural RSV infection. STUDY DESIGN A prospective cohort study was conducted at 2 academic medical centers between September 20, 2023 and March 21, 2024, enrolling 124 individuals who received the respiratory syncytial virus vaccine during pregnancy. Infant capillary blood was collected at 2 months of age from 29 of the infants. Maternal and cord immunoglobulin G levels achieved by respiratory syncytial virus vaccination were compared to those associated with maternal natural respiratory syncytial virus infection, using banked blood from 20 maternal:cord dyads collected prior to the availability of the maternal respiratory syncytial virus vaccine. Levels of immunoglobulin G against respiratory syncytial virus strain A2 and B fusion (F) and attachment (G) proteins and against pertussis toxin (as a comparator antigen from a vaccine routinely administered earlier in pregnancy) were measured using a Binding Antibody Multiplex Assay. Differences in titers between vaccination and natural infection were examined using Wilcoxon rank-sum test. Differences in cord:maternal transfer ratios and 2-month infant antibody levels by timing of maternal vaccination were evaluated by Kruskal-Wallis testing. RESULTS Maternal respiratory syncytial virus vaccination resulted in significantly higher maternal and cord antirespiratory syncytial virus F antibody levels than natural infection (5.72 vs 4.82 log10 mean fluorescence intensity, P<.0001 maternal; 5.81 vs 5.03 log10 mean fluorescence intensity, P<.0001 cord). Maternal vaccination 2 to 3 weeks and 3 to 4 weeks prior to delivery was associated with significantly lower cord:maternal transfer ratios than were observed when vaccination occurred >5 weeks prior to delivery (P=.03 for 2-3 weeks, P=.007 for 3-4 weeks), and significantly lower transfer ratios than observed for pertussis vaccination administered prior to 30 weeks of gestation (P=.008 for 2-3 weeks, P=.03 for 3-4 weeks, similar at >4 weeks). CONCLUSION Vaccine administration earlier in the approved 32 to 36 week window (at least 5 weeks prior to delivery) results in the highest transplacental transfer of maternal antibodies to the neonate. These results should inform the counseling of pregnant individuals on optimal vaccination timing.
Collapse
Affiliation(s)
- Olyvia J Jasset
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA
| | - Paola Andrea Lopez Zapana
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Zeynep Bahadir
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Lydia Shook
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA; Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | - Maria Dennis
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Emily Gilbert
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA
| | - Zhaojing Ariel Liu
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA
| | - Rachel V Yinger
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA
| | - Caroline Bald
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA
| | - Caroline G Bradford
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA
| | - Alexa H Silfen
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Sallie Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Liza Konnikova
- Departments of Pediatrics, Immunobiology, and Obstetrics, Gynecology and Reproductive Sciences, Yale University, New Haven, CT
| | - Lael M Yonker
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA
| | - Douglas Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Ashley Nelson
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Michal A Elovitz
- Women's Biomedical Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Obstetrics, Gynecology and Reproductive Sciences, Icahn school of Medicine at Mount Sinai, New York, NY
| | - Andrea G Edlow
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA; Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA.
| |
Collapse
|
6
|
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
|
7
|
Coler C, King-Nakaoka E, Every E, Chima S, Vong A, Del Rosario B, VanAbel R, Adams Waldorf KM. Impact of Infections During Pregnancy on Transplacental Antibody Transfer. Vaccines (Basel) 2024; 12:1199. [PMID: 39460363 PMCID: PMC11512415 DOI: 10.3390/vaccines12101199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/16/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024] Open
Abstract
Vaccination in pregnancy is important to protect the mother and fetus from infectious diseases. The transfer of maternal antibodies across the placenta during pregnancy can continue to protect the neonate for several months after birth while the neonatal adaptive immune system develops. Several pathogens have been shown to impair the transplacental transfer of maternal antibodies, including human immunodeficiency virus, malaria, the severe acute respiratory syndrome coronavirus 2, and cytomegalovirus. This review discusses the mechanisms contributing to decreased transplacental antibody transfer in the setting of maternal infections, such as changes in antibody glycosylation profile, maternal hypergammaglobulinemia, and placental injury. The frequency of epidemics is increasing, and pregnant people are more likely to become exposed to novel pathogens now than they were in the past. Understanding the mechanisms by which infectious diseases impair maternal-fetal antibody transfer is important for pandemic preparedness to maximize the impact of maternal vaccination for child health.
Collapse
Affiliation(s)
- Celeste Coler
- School of Medicine, University of Washington, Seattle, WA 98195, USA; (C.C.); (E.K.-N.); (E.E.)
| | - Elana King-Nakaoka
- School of Medicine, University of Washington, Seattle, WA 98195, USA; (C.C.); (E.K.-N.); (E.E.)
| | - Emma Every
- School of Medicine, University of Washington, Seattle, WA 98195, USA; (C.C.); (E.K.-N.); (E.E.)
| | - Sophia Chima
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98109, USA; (S.C.); (A.V.); (B.D.R.)
- Department of Global Health, University of Washington, Seattle, WA 98105, USA
| | - Ashley Vong
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98109, USA; (S.C.); (A.V.); (B.D.R.)
| | - Briana Del Rosario
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98109, USA; (S.C.); (A.V.); (B.D.R.)
| | - Roslyn VanAbel
- College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Kristina M. Adams Waldorf
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98109, USA; (S.C.); (A.V.); (B.D.R.)
- Department of Global Health, University of Washington, Seattle, WA 98105, USA
| |
Collapse
|
8
|
Pérez-Latorre L, Ramilo O. SARS-CoV-2 infection vs. vaccination during pregnancy: the placenta leads the way. Curr Opin Infect Dis 2024; 37:402-406. [PMID: 39105669 DOI: 10.1097/qco.0000000000001050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
PURPOSE OF REVIEW To understand the characteristics and determinants of transplacental antibody transfer against SARS-CoV-2 and to compare the differences between SARS-CoV-2 infection and vaccination. RECENT FINDINGS The need for information during the COVID-19 pandemic and the exclusion of pregnant women from randomized clinical trials have led to a vast amount of clinical data primarily based on observational studies with diverse design and sample analyses that yield variable results. This review aims to critically and comprehensively integrate the relevant knowledge related to transplacental transfer of antibodies against SARS-CoV-2, emphasizing the differences between infection and vaccination. SUMMARY Passive immunization is key to conferring protection to the infant during their first months of life. Understanding the mechanisms of transplacental antibody transfer during SARS-CoV-2 infection and vaccination, and their associated protection will allow optimizing the implementation of well tolerated and effective preventive strategies for both pregnant women and infants.
Collapse
Affiliation(s)
- Leire Pérez-Latorre
- Department of Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañon, Madrid
- CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Spain
| | - Octavio Ramilo
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
9
|
Pons-Tomàs G, Martínez-de-Albeniz I, Ríos-Barnés M, Gamell A, Simó-Nebot S, Balsells-Mejía S, Hernández-García M, Melé-Casas M, Sánchez E, Monsonis M, Gené A, López M, Salvia D, Garcia-García JJ, Fortuny C, Fumadó V. Serological Outcome in the First Months of Life of Children Born to Mothers with SARS-CoV-2 Infection during Pregnancy. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1095. [PMID: 39334627 PMCID: PMC11430284 DOI: 10.3390/children11091095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/29/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND The objective of this study is to analyze the transplacental transmission of SARS-CoV-2 antibodies, their persistence in newborns, the factors that may influence this transmission, and the protection these antibodies confer over time. METHODS This prospective cohort was conducted in a tertiary pediatric hospital in the Barcelona Metropolitan Region, Spain. It included neonates born to mothers who had SARS-CoV-2 infection during pregnancy or delivery between August 2020 and January 2022. We followed the recruited children for at least six months, and blood tests were performed to determine the presence of SARS-CoV-2 antibodies. RESULTS A total of 101 children were recruited. Among the serologies performed on children under three months of age, 44/82 were positive (53.7%). Newborns whose mothers presented more severe disease exhibited higher seropositivity odds (coefficient 9.747; p = 0.002). There were increased preterm deliveries when maternal infection occurred closer to the time of delivery. No severe SARS-CoV-2 infections were detected in children during the follow-up. CONCLUSIONS Slightly more than half of the SARS-CoV-2 serologies performed in the first three months were positive. This appears to confer protection during early childhood. The severity of maternal infection is the most significant factor influencing the transmission of antibodies in children born to unvaccinated mothers.
Collapse
Affiliation(s)
- Gemma Pons-Tomàs
- Pediatric Department, Hospital Sant Joan de Déu, University of Barcelona, 08950 Barcelona, Spain
- Infectious Diseases and Microbiome, Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Barcelona, Spain
| | | | - María Ríos-Barnés
- Infectious Diseases and Microbiome, Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Barcelona, Spain
- Infectious and Imported Diseases Department, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Anna Gamell
- Infectious Diseases and Microbiome, Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Barcelona, Spain
- Infectious and Imported Diseases Department, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Sílvia Simó-Nebot
- Infectious Diseases and Microbiome, Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Barcelona, Spain
- Infectious and Imported Diseases Department, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
| | - Sol Balsells-Mejía
- Research Promotion and Management Department, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - María Hernández-García
- Pediatric Department, Hospital Sant Joan de Déu, University of Barcelona, 08950 Barcelona, Spain
- Infectious Diseases and Microbiome, Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Barcelona, Spain
| | - Maria Melé-Casas
- Pediatric Department, Hospital Sant Joan de Déu, University of Barcelona, 08950 Barcelona, Spain
- Infectious Diseases and Microbiome, Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Barcelona, Spain
| | - Emilia Sánchez
- Blanquerna School of Health Sciences, Universitat Ramon Llull, 08022 Barcelona, Spain
| | - Manuel Monsonis
- Department of Microbiology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Amadeu Gené
- Department of Microbiology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Marta López
- Neonatology Service, Hospital Clinic La Maternitat-BCNatal Research, 08028 Barcelona, Spain
| | - Dolors Salvia
- Neonatology Service, Hospital Clinic La Maternitat-BCNatal Research, 08028 Barcelona, Spain
| | - Juan-José Garcia-García
- Pediatric Department, Hospital Sant Joan de Déu, University of Barcelona, 08950 Barcelona, Spain
- Infectious Diseases and Microbiome, Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Barcelona, Spain
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Department of Surgery and Medical-Surgical Specialties, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Claudia Fortuny
- Infectious Diseases and Microbiome, Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Barcelona, Spain
- Infectious and Imported Diseases Department, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Department of Surgery and Medical-Surgical Specialties, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Victoria Fumadó
- Infectious Diseases and Microbiome, Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Barcelona, Spain
- Infectious and Imported Diseases Department, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
- Department of Surgery and Medical-Surgical Specialties, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
10
|
Lee HY, Nazmul T, Lan J, Oyoshi MK. Maternal influences on offspring food allergy. Immunol Rev 2024; 326:130-150. [PMID: 39275992 PMCID: PMC11867100 DOI: 10.1111/imr.13392] [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: 09/16/2024]
Abstract
The prevalence of allergies has been globally escalating. While allergies could appear at any age, they often develop in early life. However, the significant knowledge gap in the field is the mechanisms by which allergies affect certain people but not others. Investigating early factors and events in neonatal life that have a lasting impact on determining the susceptibilities of children to develop allergies is a significant area of the investigation as it promotes the understanding of neonatal immune system that mediates tolerance versus allergies. This review focuses on the research over the recent 10 years regarding the potential maternal factors that influence offspring allergies with a view to food allergy, a potentially life-threatening cause of anaphylaxis. The role of breast milk, maternal diet, maternal antibodies, and microbiota that have been suggested as key maternal factors regulating offspring allergies are discussed here. We also suggest future research area to expand our knowledge of maternal-offspring interactions on the pathogenesis of food allergy.
Collapse
Affiliation(s)
- Hwa Yeong Lee
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Tanuza Nazmul
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
| | - Jinggang Lan
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
| | - Michiko K. Oyoshi
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
Okuducu YK, Mall MA, Yonker LM. COVID-19 in Pediatric Populations. Clin Chest Med 2024; 45:675-684. [PMID: 39069330 DOI: 10.1016/j.ccm.2024.02.019] [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: 07/30/2024]
Abstract
The COVID-19 pandemic reshaped the landscape of respiratory viral illnesses, causing common viruses to fade as SARS-CoV-2 took precedence. By 2023, more than 96% of the children in the United States were estimated to have been infected with SARS-CoV-2, with certain genetic predispositions and underlying health conditions posing risk factors for severe disease in children. Children, in general though, exhibit immunity advantages, protecting against aspects of the SARS-CoV-2 infection known to drive increased severity in older adults. Post-COVID-19 complications such as multisystem inflammatory syndrome in children and long COVID have emerged, underscoring the importance of vaccination. Here, we highlight the risks of severe pediatric COVID-19, age-specific immunoprotection, comparisons of SARS-CoV-2 with other respiratory viruses, and factors contributing to post-COVID-19 complications in children.
Collapse
Affiliation(s)
- Yanki K Okuducu
- Department of Pediatrics, Pulmonary Division, Massachusetts General Hospital, 175 Cambridge Street, 5(th) floor, Boston, MA 02114, USA; Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Marcus A Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin Augustenburger Platz 1, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 13353, Germany; German Center for Lung Research (DZL), Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Lael M Yonker
- Department of Pediatrics, Pulmonary Division, Massachusetts General Hospital, 175 Cambridge Street, 5(th) floor, Boston, MA 02114, USA; Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
12
|
Li L, Matsui Y, Prahl MK, Cassidy AG, Golan Y, Jigmeddagva U, Ozarslan N, Lin CY, Buarpung S, Gonzalez VJ, Chidboy MA, Basilio E, Lynch KL, Song D, Jegatheesan P, Rai DS, Govindaswami B, Needens J, Rincon M, Myatt L, Taha TY, Montano M, Ott M, Greene WC, Gaw SL. Neutralizing and binding antibody responses to SARS-CoV-2 with hybrid immunity in pregnancy. NPJ Vaccines 2024; 9:156. [PMID: 39191763 PMCID: PMC11349990 DOI: 10.1038/s41541-024-00948-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Abstract
Hybrid immunity against SARS-CoV-2 has not been well studied in pregnancy. We conducted a comprehensive analysis of neutralizing antibodies (nAb) and binding antibodies in pregnant individuals who received mRNA vaccination, natural infection, or both. A third vaccine dose augmented nAb levels compared to the two-dose regimen or natural infection alone; this effect was more pronounced in hybrid immunity. There was reduced anti-Omicron nAb, but the maternal-fetal transfer efficiency remained comparable to that of other variants. Vaccine-induced nAbs were transferred more efficiently than infection-induced nAbs. Anti-spike receptor binding domain (RBD) IgG was associated with nAb against wild-type (Wuhan-Hu-1) following breakthrough infection. Both vaccination and infection-induced anti-RBD IgA, which was more durable than anti-nucleocapsid IgA. IgA response was attenuated in pregnancy compared to non-pregnant controls. These data provide additional evidence of augmentation of humoral immune responses in hybrid immunity in pregnancy.
Collapse
Affiliation(s)
- Lin Li
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Yusuke Matsui
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Mary K Prahl
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- Division of Pediatric Infectious Diseases and Global Health, University of California San Francisco, San Francisco, CA, USA
| | - Arianna G Cassidy
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Yarden Golan
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Unurzul Jigmeddagva
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Nida Ozarslan
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Christine Y Lin
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Sirirak Buarpung
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Veronica J Gonzalez
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Megan A Chidboy
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Emilia Basilio
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Kara L Lynch
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Dongli Song
- Division of Neonatology, Department of Pediatrics, Santa Clara Valley Medical Center, San Jose, CA, USA
| | - Priya Jegatheesan
- Division of Neonatology, Department of Pediatrics, Santa Clara Valley Medical Center, San Jose, CA, USA
| | - Daljeet S Rai
- Stanford-O'Connor Family Medicine Residency Program, Division of Family Medicine, Stanford University, Palo Alto, CA, USA
| | - Balaji Govindaswami
- Division of Neonatology, Department of Pediatrics, Marshall University Joan C Edwards School of Medicine, Huntington, WV, USA
| | - Jordan Needens
- Department of Obstetrics and Gynecology, Marshall University Joan C Edwards School of Medicine, Huntington, WV, USA
| | - Monica Rincon
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, USA
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, USA
| | - Taha Y Taha
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Mauricio Montano
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Melanie Ott
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Departments of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Warner C Greene
- Gladstone Institute of Virology, San Francisco, CA, USA.
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA.
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
- Departments of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA.
| | - Stephanie L Gaw
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA.
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
13
|
Cherayil BJ, Jain N. From Womb to World: Exploring the Immunological Connections between Mother and Child. Immunohorizons 2024; 8:552-562. [PMID: 39172025 PMCID: PMC11374749 DOI: 10.4049/immunohorizons.2400032] [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: 03/20/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024] Open
Abstract
Mother and child are immunologically interconnected by mechanisms that we are only beginning to understand. During pregnancy, multiple molecular and cellular factors of maternal origin are transferred across the placenta and influence the development and function of the fetal and newborn immune system. Altered maternal immune states arising from pregnancy-associated infections or immunizations have the potential to program offspring immune function in ways that may have long-term health consequences. In this study, we review current literature on the impact of prenatal infection and vaccination on the developing immune system, highlight knowledge gaps, and look to the horizon to envision maternal interventions that could benefit both the mother and her child.
Collapse
Affiliation(s)
- Bobby J. Cherayil
- Mucosal Immunology and Biology Research Center, Mass General for Children, Charlestown, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Nitya Jain
- Mucosal Immunology and Biology Research Center, Mass General for Children, Charlestown, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
- Center for Computational and Integrative Biology, Mass General Brigham, Boston, MA
| |
Collapse
|
14
|
Martínez-Quezada R, Valencia-Ledezma OE, Ramírez-Lozada T, Miguel-Rodríguez CE, Fernández-Hernández JC, Acosta-Altamirano G. Influence of Maternal and Neonatal Factors on Transplacental Passive Immunity after Vaccination against COVID-19. Vaccines (Basel) 2024; 12:860. [PMID: 39203986 PMCID: PMC11360686 DOI: 10.3390/vaccines12080860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/20/2024] [Accepted: 07/29/2024] [Indexed: 09/03/2024] Open
Abstract
In the present study, we evaluated the influence of maternal and neonatal factors on the efficiency of the placental transfer of neutralizing antibodies against SARS-CoV-2. Vaccination during pregnancy provides fetal and neonatal protection through the passive transplacental transfer of maternal neutralizing antibodies. To date, little information is available regarding the factors that affect the transfer of antibodies against SARS-CoV-2. A retrospective, cross-sectional, observational, and analytical study was carried out. It was found that several biological factors could be altering transplacental passive immunity after vaccination against COVID-19. In our study population, type 2 diabetes mellitus and chronic hypertension tended to decrease efficiency, while data from women with pre-eclampsia showed better indices compared to those from women with healthy pregnancies. Neonates born prematurely showed lower transfer rates when compared to healthy neonates. The premature rupture of membranes significantly decreased antibody transfer. Taken together, the data suggest that vaccination against COVID-19 during pregnancy is effective even under certain unfavorable clinical conditions for the mother, fetus, and neonate. It is important to create and disseminate immunization strategies in vulnerable populations to reduce maternal and perinatal morbidity and mortality associated with infections preventable by vaccination.
Collapse
Affiliation(s)
- Rebeca Martínez-Quezada
- Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, IMSS-Bienestar, Ixtapaluca 56530, Mexico; (R.M.-Q.); (O.E.V.-L.); (C.E.M.-R.)
- Consejo Mexiquense de Ciencia y Tecnología (COMECYT), Toluca 50120, Mexico
| | - Omar Esteban Valencia-Ledezma
- Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, IMSS-Bienestar, Ixtapaluca 56530, Mexico; (R.M.-Q.); (O.E.V.-L.); (C.E.M.-R.)
| | - Tito Ramírez-Lozada
- Servicio de Ginecología y Obstetricia, Hospital Regional de Alta Especialidad de Ixtapaluca, IMSS-Bienestar, Ixtapaluca 56530, Mexico;
| | - Carlos Emilio Miguel-Rodríguez
- Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, IMSS-Bienestar, Ixtapaluca 56530, Mexico; (R.M.-Q.); (O.E.V.-L.); (C.E.M.-R.)
| | | | - Gustavo Acosta-Altamirano
- Dirección de Investigación, Hospital General de México “Eduardo Liceaga”, Ciudad de México 06726, Mexico;
| |
Collapse
|
15
|
Jasset MOJ, Andrea Lopez Zapana P, Bahadir Z, Shook L, Dennis MM, Gilbert ME, Liu MZA, Yinger MRV, Bald MC, Bradford MCG, Silfen MAH, Klein SL, Pekosz A, Permar S, Konnikova L, Yonker LM, Lauffenburger D, Nelson A, Elovitz MA, Edlow AG. Longer interval between maternal RSV vaccination and birth increases placental transfer efficiency. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.14.24310390. [PMID: 39072025 PMCID: PMC11275666 DOI: 10.1101/2024.07.14.24310390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Background Respiratory Syncytial Virus (RSV) is associated with significant neonatal and infant morbidity and mortality. Maternal bivalent RSVpreF RSV vaccination to protect neonates and infants was approved in September 2023 for administration between 32+0 and 36+6 weeks to protect neonates and infants. This approved timeframe is narrower than the 24-36 week window evaluated in the clinical trial, due to the possible association between preterm birth and vaccine administration. Currently, data are lacking on how maternal vaccine timing within the approved window affects the transfer of antibodies from mother to fetus, critical information that could influence clinical practice. Objectives We sought to examine how gestational age at vaccination and time elapsed from maternal RSV vaccination to delivery impacted transfer of maternal antibodies measured in the umbilical cord at delivery and in peripheral blood of 2-month infants. We also examined differences in maternal and cord RSV antibody levels achieved by vaccination versus natural RSV infection. Study Design A prospective cohort study was conducted at two academic medical centers between September 20, 2023 and March 21, 2024, enrolling 124 individuals who received the RSV vaccine during pregnancy. Infant capillary blood was collected at 2 months of age from 29 of the infants. Maternal and cord IgG levels achieved by RSV vaccination were compared to those associated with maternal natural RSV infection, using banked blood from 20 maternal:cord dyads collected prior to the availability of the maternal RSV vaccine. Levels of IgG against RSV strain A2 and B fusion (F) and attachment (G) proteins and against pertussis toxin (as a comparator antigen from a vaccine routinely administered earlier in pregnancy) were measured using a Binding Antibody Multiplex Assay. Differences in titers between vaccination and natural infection were examined using Wilcoxon rank sum test. Differences in cord:maternal transfer ratios and 2-month infant antibody levels by timing of maternal vaccination were evaluated by Kruskal-Wallis testing. Results Maternal RSV vaccination resulted in significantly higher maternal and cord anti-F RSV antibody levels than natural infection (5.72 vs 4.82 log 10 MFI, p < 0.0001 maternal; 5.81 vs 5.03 log 10 MFI, p < 0.0001 cord). Maternal vaccination 2-3 weeks and 3-4 weeks prior to delivery was associated with significantly lower cord:maternal transfer ratios than were observed when vaccination occurred > 5 weeks prior to delivery (p = 0.03 for 2-3 weeks, p = 0.007 for 3-4 weeks), and significantly lower transfer ratios than observed for pertussis vaccination administered prior to 30 weeks' gestation (p = 0.008 for 2-3 weeks, p = 0.03 for 3-4 weeks, similar at > 4 weeks). Conclusions Vaccine administration earlier in the approved 32-36 week window (at least 5 weeks prior to delivery) results in the highest transplacental transfer of maternal antibodies to the neonate. These results should inform the counseling of pregnant individuals on optimal vaccination timing.
Collapse
|
16
|
Costa S, Giordano L, Bottoni A, Tiberi E, Fattore S, Pastorino R, Simone ND, Lanzone A, Buonsenso D, Valentini P, Cattani P, Santangelo R, Sanguinetti M, Scambia G, Vento G. Vertical Transmission of SARS-CoV-2 during Pregnancy: A Prospective Italian Cohort Study. Am J Perinatol 2024; 41:1077-1085. [PMID: 35263767 DOI: 10.1055/a-1792-4535] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The extent of vertical transmission (VT) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) from mothers their fetuses or neonates is still uncertain. We aimed to determine the incidence of VT. STUDY DESIGN In this prospective cohort study. All mother diagnosed with SARS-CoV-2 infection at the time of delivery or up to 1 week prior and their neonates, managed in a tertiary referral hospital for pregnancy complicated by coronavirus disease 2019 (COVID-19) in Rome, from April 2 to December 22, 2020, were included. Maternal infection was defined as nasopharyngeal swab test results positive for SARS-CoV-2 reverse transcription-polymerase chain reaction (RT-PCR). Biological samples were collected before, at, and after delivery to test positivity for SARS-CoV-2 RT-PCR and anti-SARS-CoV-2-specific antibodies. RESULTS The cohort included 95 women and 96 neonates with documented SARS-CoV-2 test results. Four neonates (4.2%) tested positive. The incidence of VT, according to the guidance criteria for diagnosing perinatal SARS-CoV-2 infection, was 5.2%. Neonatal symptoms were due to prematurity or fetal distress: symptomatic infants had lower median (min-max) gestational age, 38.1 (29.3-40.6) versus 39.3 (33.9-41.9) weeks (p = 0.036), and 1-minute and 5-minute Apgar scores, 9 (3-9) versus 9 (7-10) (p = 0.036) and 10 (6-10) versus 10 (8-10) (p = 0.012), respectively, than asymptomatic infants and needed more frequent assistance in the delivery room (22.2 vs 2.5%; p = 0.008). Only six (7.1%) neonates had anti-SARS-CoV-2-specific antibodies, despite the ongoing maternal infection. CONCLUSION The incidence of VT is low as is the detection of specific anti-SARS-CoV-2 antibodies in cord blood when infection is contracted late in pregnancy. This would suggest poor protection of infants against horizontal transmission of the virus. KEY POINTS · VT of SARS-CoV-2 from pregnant mothers to fetuses or neonates can be possible.. · In this prospective cohort study, the incidence of VT is found to be 5.2%.. · VT is low but exists..
Collapse
Affiliation(s)
- Simonetta Costa
- Department of Woman and Child Health and Public Health, Catholic University of Sacred Heart, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Lucia Giordano
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Anthea Bottoni
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Eloisa Tiberi
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Simona Fattore
- Department of Woman and Child Health and Public Health, Catholic University of Sacred Heart, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Roberta Pastorino
- Department of Woman and Child Health and Public Health, Catholic University of Sacred Heart, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Nicoletta Di Simone
- Department of Biomedical Sciences, Humanitas University, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Antonio Lanzone
- Department of Woman and Child Health and Public Health, Catholic University of Sacred Heart, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Danilo Buonsenso
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Piero Valentini
- Department of Woman and Child Health and Public Health, Catholic University of Sacred Heart, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Paola Cattani
- Department of Woman and Child Health and Public Health, Catholic University of Sacred Heart, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Rosaria Santangelo
- Department of Laboratory and Infectivology Science, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of Sacred Heart, Rome, Italy
| | - Maurizio Sanguinetti
- Department of Laboratory and Infectivology Science, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of Sacred Heart, Rome, Italy
| | - Giovanni Scambia
- Department of Woman and Child Health and Public Health, Catholic University of Sacred Heart, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giovanni Vento
- Department of Woman and Child Health and Public Health, Catholic University of Sacred Heart, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
17
|
Vercoutere A, Zina MJ, Telis M, Goffard JC, Boulvain M, de Doncker L, Derisbourg S, Houben S, Delforge ML, Daelemans C, Kelen D. Seroprevalence and placental transfer of SARS-CoV-2 antibodies in unvaccinated pregnant women. BMC Infect Dis 2024; 24:509. [PMID: 38773493 PMCID: PMC11110414 DOI: 10.1186/s12879-024-09399-6] [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] [Received: 12/18/2023] [Accepted: 05/13/2024] [Indexed: 05/23/2024] Open
Abstract
PURPOSE Pregnant women are at risk of severe SARS-CoV-2 infection, potentially leading to obstetric and neonatal complications. Placental transfer of antibodies directed to SARS-CoV-2 may be protective against neonatal COVID-19, but this remains to be studied. We aimed to determine the seroprevalence of SARS-CoV-2 antibodies in a population of unvaccinated pregnant women and to determine the placental transfer of these antibodies. METHODOLOGY A total of 1197 unvaccinated women with mostly unknown pre-study SARS-CoV-2 infection status, were tested at delivery for SARS-CoV-2 spike protein IgG antibodies during the first year of the pandemic. Umbilical cord samples were collected and assessed for seropositivity if the mother was seropositive. Maternal characteristics, pregnancy and neonatal outcomes and data on SARS-CoV-2 infection were extracted from medical records. RESULTS Specific IgG were detected in 258 women (21.6%). A significant placental transfer to the newborn was observed in 81.3% of cases. The earlier in the 2nd and 3rd trimesters that the mother had contracted the disease and the more symptomatic she was, the greater the likelihood of transplacental transfer of IgG to her newborn. CONCLUSION Approximately one in five women had detectable anti-SARS-CoV-2 spike protein IgG antibodies at delivery during the first year of the pandemic, and these antibodies were significantly transferred to their fetuses. This research provides further evidence to better understand the dynamics of the placental transfer of SARS-CoV-2 IgG antibodies from mothers to their newborns, which is necessary to improve vaccination strategies.
Collapse
Affiliation(s)
- An Vercoutere
- Department of Obstetrics and Gynaecology, Hôpital Erasme, Hôpital Universitaire de Bruxelles (H.U.B.), Route de Lennik 808, 1070, Brussels, Belgium.
| | | | - Meltem Telis
- Department of Neonatology, Hôpital Erasme, Hôpital Universitaire de Bruxelles (H.U.B.), Brussels, Belgium
| | - Jean-Christophe Goffard
- Department of Internal Medicine, Hôpital Erasme, Hôpital Universitaire de Bruxelles (H.U.B.), Brussels, Belgium
| | - Michel Boulvain
- Department of Obstetrics and Gynaecology, Hôpital Erasme, Hôpital Universitaire de Bruxelles (H.U.B.), Route de Lennik 808, 1070, Brussels, Belgium
| | - Loïc de Doncker
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Sara Derisbourg
- Department of Obstetrics and Gynaecology, Hôpital Erasme, Hôpital Universitaire de Bruxelles (H.U.B.), Route de Lennik 808, 1070, Brussels, Belgium
| | - Sylvie Houben
- Department of Obstetrics and Gynaecology, Hôpital Delta, Chirec Hospitals, Brussels, Belgium
| | - Marie-Luce Delforge
- National Reference Center for Congenital Infections, Hôpital Erasme, Hôpital Universitaire de Bruxelles (H.U.B.), Brussels, Belgium
| | - Caroline Daelemans
- Department of Woman, Child and Adolescent Medicine, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Dorottya Kelen
- Department of Neonatology, Hôpital Erasme, Hôpital Universitaire de Bruxelles (H.U.B.), Brussels, Belgium
| |
Collapse
|
18
|
Fernandes KA, Lim AI. Maternal-driven immune education in offspring. Immunol Rev 2024; 323:288-302. [PMID: 38445769 DOI: 10.1111/imr.13315] [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: 03/07/2024]
Abstract
Maternal environmental exposures, particularly during gestation and lactation, significantly influence the immunological development and long-term immunity of offspring. Mammalian immune systems develop through crucial inputs from the environment, beginning in utero and continuing after birth. These critical developmental windows are essential for proper immune system development and, once closed, may not be reopened. This review focuses on the mechanisms by which maternal exposures, particularly to pathogens, diet, and microbiota, impact offspring immunity. Mechanisms driving maternal-offspring immune crosstalk include transfer of maternal antibodies, changes in the maternal microbiome and microbiota-derived metabolites, and transfer of immune cells and cytokines via the placenta and breastfeeding. We further discuss the role of transient maternal infections, which are common during pregnancy, in providing tissue-specific immune education to offspring. We propose a "maternal-driven immune education" hypothesis, which suggests that offspring can use maternal encounters that occur during a critical developmental window to develop optimal immune fitness against infection and inflammation.
Collapse
Affiliation(s)
| | - Ai Ing Lim
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| |
Collapse
|
19
|
Ghosh R, Gutierrez JP, de Jesús Ascencio-Montiel I, Juárez-Flores A, Bertozzi SM. SARS-CoV-2 infection by trimester of pregnancy and adverse perinatal outcomes: a Mexican retrospective cohort study. BMJ Open 2024; 14:e075928. [PMID: 38604636 PMCID: PMC11015228 DOI: 10.1136/bmjopen-2023-075928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 03/04/2024] [Indexed: 04/13/2024] Open
Abstract
OBJECTIVE Conflicting evidence for the association between COVID-19 and adverse perinatal outcomes exists. This study examined the associations between maternal COVID-19 during pregnancy and adverse perinatal outcomes including preterm birth (PTB), low birth weight (LBW), small-for-gestational age (SGA), large-for-gestational age (LGA) and fetal death; as well as whether the associations differ by trimester of infection. DESIGN AND SETTING The study used a retrospective Mexican birth cohort from the Instituto Mexicano del Seguro Social (IMSS), Mexico, between January 2020 and November 2021. PARTICIPANTS We used the social security administrative dataset from IMSS that had COVID-19 information and linked it with the IMSS routine hospitalisation dataset, to identify deliveries in the study period with a test for SARS-CoV-2 during pregnancy. OUTCOME MEASURES PTB, LBW, SGA, LGA and fetal death. We used targeted maximum likelihood estimators, to quantify associations (risk ratio, RR) and CIs. We fit models for the overall COVID-19 sample, and separately for those with mild or severe disease, and by trimester of infection. Additionally, we investigated potential bias induced by missing non-tested pregnancies. RESULTS The overall sample comprised 17 340 singleton pregnancies, of which 30% tested positive. We found that those with mild COVID-19 had an RR of 0.89 (95% CI 0.80 to 0.99) for PTB and those with severe COVID-19 had an RR of 1.53 (95% CI 1.07 to 2.19) for LGA. COVID-19 in the first trimester was associated with fetal death, RR=2.36 (95% CI 1.04, 5.36). Results also demonstrate that missing non-tested pregnancies might induce bias in the associations. CONCLUSIONS In the overall sample, there was no evidence of an association between COVID-19 and adverse perinatal outcomes. However, the findings suggest that severe COVID-19 may increase the risk of some perinatal outcomes, with the first trimester potentially being a high-risk period.
Collapse
Affiliation(s)
- Rakesh Ghosh
- Institute for Global Health Sciences, University of California San Francisco, San Francisco, California, USA
- School of Public Health, University of California Berkeley, Berkeley, California, USA
| | - Juan Pablo Gutierrez
- Center for Policy, Population & Health Research, National Autonomous University of Mexico, Mexico City, Mexico
| | | | - Arturo Juárez-Flores
- Center for Policy, Population & Health Research, National Autonomous University of Mexico, Mexico City, Mexico
| | - Stefano M Bertozzi
- School of Public Health, University of California Berkeley, Berkeley, California, USA
- University of Washington - Seattle Campus, Seattle, Washington, USA
- National Institute of Public Health, Cuernavaca, Mexico
| |
Collapse
|
20
|
Lopez PA, Nziza N, Chen T, Shook LL, Burns MD, Demidkin S, Jasset O, Akinwunmi B, Yonker LM, Gray KJ, Elovitz MA, Lauffenburger DA, Julg BD, Edlow AG. Placental transfer dynamics and durability of maternal COVID-19 vaccine-induced antibodies in infants. iScience 2024; 27:109273. [PMID: 38444609 PMCID: PMC10914478 DOI: 10.1016/j.isci.2024.109273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/18/2024] [Accepted: 02/15/2024] [Indexed: 03/07/2024] Open
Abstract
Completion of a COVID-19 vaccination series during pregnancy effectively reduces COVID-19 hospitalization among infants less than 6 months of age. The dynamics of transplacental transfer of maternal vaccine-induced antibodies, and their persistence in infants at 2, 6, 9, and 12 months, have implications for new vaccine development and optimal timing of vaccine administration in pregnancy. We evaluated anti-COVID antibody IgG subclass, Fc-receptor binding profile, and activity against wild-type Spike and RBD plus five variants of concern (VOCs) in 153 serum samples from 100 infants. Maternal IgG1 and IgG3 responses persisted in 2- and 6-month infants to a greater extent than the other IgG subclasses, with high persistence of antibodies binding placental neonatal Fc-receptor and FcγR3A. Lowest persistence was observed against the Omicron RBD-specific region. Maternal vaccine timing, placental Fc-receptor binding capabilities, antibody subclass, fetal sex, and VOC all impact the persistence of antibodies in infants through 12 months of age.
Collapse
Affiliation(s)
- Paola A. Lopez
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Nadège Nziza
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Tina Chen
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Lydia L. Shook
- Massachusetts General Hospital, Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Boston, MA 02114, USA
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Madeleine D. Burns
- Massachusetts General Hospital for Children, Department of Pediatric, Boston, MA 02114, USA
| | - Stepan Demidkin
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA 02114, USA
| | - Olyvia Jasset
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA 02114, USA
| | - Babatunde Akinwunmi
- Brigham and Women’s Hospital, Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Boston, MA 02115, USA
| | - Lael M. Yonker
- Harvard Medical School, Boston, MA 02115, USA
- Massachusetts General Hospital for Children, Department of Pediatric, Boston, MA 02114, USA
| | - Kathryn J. Gray
- Harvard Medical School, Boston, MA 02115, USA
- Brigham and Women’s Hospital, Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Boston, MA 02115, USA
| | - Michal A. Elovitz
- Women’s Biomedical Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Douglas A. Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Boris D. Julg
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Andrea G. Edlow
- Massachusetts General Hospital, Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Boston, MA 02114, USA
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
21
|
Adhikari EH, Lu P, Kang YJ, McDonald AR, Pruszynski JE, Bates TA, McBride SK, Trank-Greene M, Tafesse FG, Lu LL. Diverging Maternal and Cord Antibody Functions From SARS-CoV-2 Infection and Vaccination in Pregnancy. J Infect Dis 2024; 229:462-472. [PMID: 37815524 PMCID: PMC10873180 DOI: 10.1093/infdis/jiad421] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/27/2023] [Accepted: 09/29/2023] [Indexed: 10/11/2023] Open
Abstract
Maternal immunity impacts the infant, but how is unclear. To understand the implications of the immune exposures of vaccination and infection in pregnancy for neonatal immunity, we evaluated antibody functions in paired peripheral maternal and cord blood. We compared those who in pregnancy received mRNA coronavirus disease 2019 (COVID-19) vaccine, were infected by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and the combination. We found that vaccination enriched a subset of neutralizing activities and Fc effector functions that was driven by IgG1 and was minimally impacted by antibody glycosylation in maternal blood. In paired cord blood, maternal vaccination also enhanced IgG1. However, Fc effector functions compared to neutralizing activities were preferentially transferred. Moreover, changes in IgG posttranslational glycosylation contributed more to cord than peripheral maternal blood antibody functional potency. These differences were enhanced with the combination of vaccination and infection as compared to either alone. Thus, Fc effector functions and antibody glycosylation highlight underexplored maternal opportunities to safeguard newborns.
Collapse
Affiliation(s)
- Emily H Adhikari
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Parkland Health, Dallas Texas, USA
| | - Pei Lu
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ye Jin Kang
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ann R McDonald
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jessica E Pruszynski
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Timothy A Bates
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Savannah K McBride
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Mila Trank-Greene
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Fikadu G Tafesse
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Lenette L Lu
- Parkland Health, Dallas Texas, USA
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
22
|
Chiang CJ, Hsu WL, Su MT, Ko WC, Hsu KF, Tsai PY. Impact of Antenatal SARS-CoV-2 Exposure on SARS-CoV-2 Neutralization Potency. Vaccines (Basel) 2024; 12:164. [PMID: 38400147 PMCID: PMC10892697 DOI: 10.3390/vaccines12020164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/25/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
A pregnancy booster dose significantly reduces the risk and severity of COVID-19, and it is widely recommended. A prospective cohort study was conducted to compare the transplacental passage of maternal antibodies from vaccination or infection during three trimesters against both the vaccine-targeted Wuhan strain and the Omicron strain of SARS-CoV-2. Maternal-infant dyads from vaccinated mothers were collected between 6 June 2022 and 20 September 2022. We analyzed 38 maternal-infant dyads from mothers who had been infected with COVID-19 and 37 from mothers without any previous infection. Pregnant women who received their last COVID-19 vaccine dose in the third trimester exhibited the highest anti-spike protein antibody levels and neutralizing potency against both the Wuhan strain and Omicron BA.2 variant in their maternal and cord plasma. Both second- and third-trimester vaccination could lead to a higher level of neutralization against the Wuhan and Omicron strains. COVID-19 infection had a negative effect on the transplacental transfer ratio of SARS-CoV-2 antibodies. A booster dose during the second or third trimester is encouraged for the maximum transplacental transfer of humoral protection against COVID-19 for infants.
Collapse
Affiliation(s)
- Chia-Jung Chiang
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan
| | - Wei-Lun Hsu
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan
| | - Mei-Tsz Su
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan
| | - Wen-Chien Ko
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan
- Center for Infection Control, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan
| | - Keng-Fu Hsu
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan
| | - Pei-Yin Tsai
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan
| |
Collapse
|
23
|
Lauritsen CJ, Trinh IV, Desai SP, Clancey E, Murrell AE, Rambaran S, Chandra S, Elliott DH, Smira AR, Mo Z, Stone AE, Agbodji A, Dugas CM, Satou R, Pridjian G, Longo S, Ley SH, Robinson JE, Norton EB, Piedimonte G, Gunn BM. Passive antibody transfer from pregnant women to their fetus are maximized after SARS-CoV-2 vaccination irrespective of prior infection. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2024; 3:100189. [PMID: 38268538 PMCID: PMC10805668 DOI: 10.1016/j.jacig.2023.100189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/19/2023] [Accepted: 08/14/2023] [Indexed: 01/26/2024]
Abstract
Background Pregnancy is associated with a higher risk of adverse symptoms and outcomes for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection for both mother and neonate. Antibodies can provide protection against SARS-CoV-2 infection and are induced in pregnant women after vaccination or infection. Passive transfer of these antibodies from mother to fetus in utero may provide protection to the neonate against infection. However, it is unclear whether the magnitude or quality and kinetics of maternally derived fetal antibodies differs in the context of maternal infection or vaccination. Objective We aimed to determine whether antibodies transferred from maternal to fetus differed in quality or quantity between infection- or vaccination-induced humoral immune responses. Methods We evaluated 93 paired maternal and neonatal umbilical cord blood plasma samples collected between October 2020 and February 2022 from a birth cohort of pregnant women from New Orleans, Louisiana, with histories of SARS-CoV-2 infection and/or vaccination. Plasma was profiled for the levels of spike-specific antibodies and induction of antiviral humoral immune functions, including neutralization and Fc-mediated innate immune effector functions. Responses were compared between 4 groups according to maternal infection and vaccination. Results We found that SARS-CoV-2 vaccination or infection during pregnancy increased the levels of antiviral antibodies compared to naive subjects. Vaccinated mothers and cord samples had the highest anti-spike antibody levels and antiviral function independent of the time of vaccination during pregnancy. Conclusions These results show that the most effective passive transfer of functional antibodies against SARS-CoV-2 in utero is achieved through vaccination, highlighting the importance of vaccination in pregnant women.
Collapse
Affiliation(s)
- Cody J. Lauritsen
- Paul G. Allen School of Global Health, Washington State University, Pullman, Wash
| | - Ivy V. Trinh
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, La
| | - Srushti P. Desai
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Erin Clancey
- Paul G. Allen School of Global Health, Washington State University, Pullman, Wash
| | - Amelie E. Murrell
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, La
| | - Saraswatie Rambaran
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, La
| | - Sruti Chandra
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Debra H. Elliott
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Ashley R. Smira
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Zhiyin Mo
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, La
| | - Addison E. Stone
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, La
| | - Ayitevi Agbodji
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Courtney M. Dugas
- Department of Physiology, Tulane University School of Medicine, New Orleans, La
| | - Ryousuke Satou
- Department of Physiology, Tulane University School of Medicine, New Orleans, La
| | - Gabriella Pridjian
- Department of Obstetrics and Gynecology, Tulane University School of Medicine, New Orleans, La
| | | | - Sylvia H. Ley
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, La
| | - James E. Robinson
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Elizabeth B. Norton
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, La
| | - Giovanni Piedimonte
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Bronwyn M. Gunn
- Paul G. Allen School of Global Health, Washington State University, Pullman, Wash
| |
Collapse
|
24
|
Martínez-Quezada R, Miguel-Rodríguez CE, Ramírez-Lozada T, Valencia-Ledezma OE, Acosta-Altamirano G. Placental Transfer Efficiency of Neutralizing Antibodies on SARS-CoV-2 Vaccination before and after Pregnancy in Mexican Women. Int J Mol Sci 2024; 25:1516. [PMID: 38338795 PMCID: PMC10855582 DOI: 10.3390/ijms25031516] [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: 12/05/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 02/12/2024] Open
Abstract
The protection of the neonate against pathogens depends largely on the antibodies transferred placentally from the mother; for this reason, maternal vaccination against emerging viruses, such as SARS-CoV-2, is of vital importance. Knowing some of the immunogenic factors that could alter the placental transfer of antibodies could aid in understanding the immune response and neonatal protection after maternal vaccination. In this study, we analyzed the efficiency of the placental transfer of binding and neutralizing antibodies, as well as some factors that could alter the passive immune response, such as the trimester of gestation at the time of immunization, the number of doses received by the mother and the type of vaccine. Binding IgG antibodies were detected by ELISA, and the detection of neutralizing antibodies was carried out using flow cytometry. Our results show efficient transfer rates (>1), which are higher when maternal vaccination occurs during the third trimester of gestation. Antibodies are detectable in mothers and their neonates after 12 months of maternal immunization, suggesting than the vaccination against COVID-19 before and during pregnancy in the Mexican population induces a lasting neutralizing response in mothers and their newborns.
Collapse
Affiliation(s)
- Rebeca Martínez-Quezada
- Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, Carretera Federal México-Puebla Km. 34.5, Pueblo de Zoquiapan, Ixtapaluca 56530, Mexico; (R.M.-Q.); (C.E.M.-R.); (O.E.V.-L.)
- Consejo Mexiquense de Ciencia y Tecnología (COMECYT), Paseo Colón N° 112-A, Ciprés, Toluca 50120, Mexico
| | - Carlos Emilio Miguel-Rodríguez
- Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, Carretera Federal México-Puebla Km. 34.5, Pueblo de Zoquiapan, Ixtapaluca 56530, Mexico; (R.M.-Q.); (C.E.M.-R.); (O.E.V.-L.)
| | - Tito Ramírez-Lozada
- Unidad de Ginecología y Obstetricia, Hospital Regional de Alta Especialidad de Ixtapaluca, Carretera Federal México-Puebla Km. 34.5, Pueblo de Zoquiapan, Ixtapaluca 56530, Mexico;
| | - Omar Esteban Valencia-Ledezma
- Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, Carretera Federal México-Puebla Km. 34.5, Pueblo de Zoquiapan, Ixtapaluca 56530, Mexico; (R.M.-Q.); (C.E.M.-R.); (O.E.V.-L.)
| | - Gustavo Acosta-Altamirano
- Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, Carretera Federal México-Puebla Km. 34.5, Pueblo de Zoquiapan, Ixtapaluca 56530, Mexico; (R.M.-Q.); (C.E.M.-R.); (O.E.V.-L.)
| |
Collapse
|
25
|
Uta M, Craina M, Marc F, Enatescu I. Assessing the Impact of COVID-19 Vaccination on Preterm Birth: A Systematic Review with Meta-Analysis. Vaccines (Basel) 2024; 12:102. [PMID: 38276674 PMCID: PMC10818953 DOI: 10.3390/vaccines12010102] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/09/2024] [Accepted: 01/14/2024] [Indexed: 01/27/2024] Open
Abstract
During the coronavirus diseases 2019 (COVID-19) pandemic, the safety and efficacy of vaccination during pregnancy, particularly regarding the risk of preterm birth, have been a subject of concern. This systematic review aims to evaluate the impact of COVID-19 vaccination on preterm birth risk and to inform clinical practice and public health policies. Following PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines, a database search included PubMed, Embase, and Scopus, conducted up until October 2023. Inclusion criteria focused on studies that examined COVID-19 vaccination during pregnancy and its correlation with preterm birth, defined as a birth before 37 weeks of gestation. Six studies met these criteria, encompassing 35,612 patients. A quality assessment was performed using the Newcastle-Ottawa Scale and the Cochrane Collaboration's tool, with the risk of bias evaluated via a funnel plot analysis and an Egger's regression test. The studies demonstrated geographical diversity, mainly from Israel, Romania, and the United States, with a blend of prospective and retrospective designs. The patient cohort's mean age was 31.2 years, with common comorbidities such as gestational diabetes and obesity affecting 9.85% of the total population. The vaccination types varied across the studies, with BNT162b2 being the most used. The results indicated a low heterogeneity among the included studies, evidenced by a Cochran's Q statistic of 2.10 and an I2 statistic of 13%. The meta-analysis yielded a pooled odds ratio (OR) for a preterm birth risk post-vaccination of approximately 1.03 (95% CI: 0.82-1.30), suggesting no significant increase in preterm birth risk was associated with COVID-19 vaccination. Notable findings included a low preterm birth rate (as low as 0.6% and up to 6.1%) with minimal differences in neonatal outcomes, such as birth weight and APGAR (appearance, pulse, grimace, activity, and respiration) scores between vaccinated and unvaccinated groups. This study concludes that a COVID-19 vaccination during pregnancy does not significantly increase the risk of preterm birth. These findings are crucial for reassuring healthcare providers and pregnant women about the safety of COVID-19 vaccines and supporting their use in public health strategies during the pandemic.
Collapse
Affiliation(s)
- Mihaela Uta
- Discipline of Obstetrics and Gynecology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (M.U.); (M.C.)
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Marius Craina
- Discipline of Obstetrics and Gynecology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (M.U.); (M.C.)
| | - Felicia Marc
- Department of Medical Sciences, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Ileana Enatescu
- Discipline of Neonatology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| |
Collapse
|
26
|
Khalifeh M, Rubin LG, Dayya D, Cerise J, Skinner K, Maloney M, Walworth C, Petropoulos CJ, Wrin T, Chun K, Weinberger B. SARS-CoV-2 neutralizing antibody titers in maternal blood, umbilical cord blood, and breast milk. J Perinatol 2024; 44:28-34. [PMID: 38092879 DOI: 10.1038/s41372-023-01843-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/16/2023] [Accepted: 11/24/2023] [Indexed: 01/13/2024]
Abstract
OBJECTIVE We quantified neutralizing SARS-CoV-2 antibody against spike protein (nAb) levels after vaccination and SARS-CoV-2 infection in maternal serum, cord blood, and breast milk and determined whether they correlate with levels of spike protein binding antibody. STUDY DESIGN Women (n = 100) were enrolled on admission for delivery. Previous SARS-CoV-2 infection was defined by anti-nucleocapsid antibodies. Levels of nAb and binding antibodies against spike receptor binding domain were measured in maternal blood, cord blood, and milk. RESULTS Maternal nAb levels were higher after vaccine and infection than vaccine alone but waned rapidly. Levels of nAb in cord blood and milk correlated with maternal levels and were higher in cord blood than maternal. Spike protein binding antibody levels correlated with nAb. CONCLUSION SARS-CoV-2 vaccination near delivery may boost antibody-mediated immunity in the peripartum period. Neutralizing antibodies are passed transplacentally and into milk. Spike protein binding antibody may be a feasible proxy for nAb.
Collapse
Affiliation(s)
- Mazen Khalifeh
- Division of Neonatology and Newborn Medicine, Phelps Hospital, Northwell Health, Sleepy Hollow, NY, USA
| | - Lorry G Rubin
- Division of Pediatric Infectious Diseases, Cohen Children's Medical Center, Northwell Health and Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, NY, USA
| | - David Dayya
- Division of Undersea and Hyperbaric Medicine, Department of Surgery, Phelps Hospital, Northwell Health, Sleepy Hollow, NY, USA
| | - Jane Cerise
- Biostatistics Unit, Office of Academic Affairs, Northwell School, New Hyde Park, NY, USA
| | - Karen Skinner
- Division of Neonatology and Newborn Medicine, Phelps Hospital, Northwell Health, Sleepy Hollow, NY, USA
| | - Mona Maloney
- Division of Neonatology and Newborn Medicine, Phelps Hospital, Northwell Health, Sleepy Hollow, NY, USA
| | - Charles Walworth
- Labcorp-Monogram Biosciences, South San Francisco, CA, 94080, USA
| | | | - Terri Wrin
- Labcorp-Monogram Biosciences, South San Francisco, CA, 94080, USA
| | - Kelly Chun
- Labcorp-Esoterix, Calabasas, CA, 91301, USA
| | - Barry Weinberger
- Division of Neonatal-Perinatal Medicine, Cohen Children's Medical Center, Northwell Health and Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, NY, USA.
| |
Collapse
|
27
|
Devera JL, Gonzalez Y, Sabharwal V. A narrative review of COVID-19 vaccination in pregnancy and breastfeeding. J Perinatol 2024; 44:12-19. [PMID: 37495712 DOI: 10.1038/s41372-023-01734-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/22/2023] [Accepted: 07/17/2023] [Indexed: 07/28/2023]
Abstract
The Coronavirus pandemic has affected millions of people due to the spread of the Severe acute respiratory syndrome Coronavirus-2 (SARS-CoV-2) virus. Pregnant individuals and infants are most vulnerable given the increased risk of developing severe complications from SARS-CoV-2 infection. Recently, COVID-19 vaccination is recommended for pregnant women and infants starting at 6 months of age to prevent disease contraction and minimize disease severity. We conducted a review of the literature on COVID-19 vaccination to discuss vaccine safety and efficacy, immunity after maternal vaccination, transplacental transfer and persistence of antibodies, and public health implications. Current evidence supports the safety and efficacy of vaccination during pregnancy. Maternal vaccination provides greater antibody persistence in infants compared to immunity from natural infection. Furthermore, vaccination has demonstrated an increased rate of passive antibody transfer through the placenta and breast milk. Public health interventions are important in achieving herd immunity and ultimately ending the pandemic. IMPACT: This article highlights the benefits of COVID-19 vaccination during pregnancy with a review of the data describing safety and efficacy, passive and active immunity after maternal immunization, trans-placental transfer and persistence of protective antibodies, and public health implications. With this information, healthcare providers can provide up-to-date knowledge to their pregnant patients to help them form an informed decision on vaccination and combat vaccine hesitancy.
Collapse
Affiliation(s)
- Jean L Devera
- Chobanian & Avedisian School of Medicine, Boston, MA, US
| | | | - Vishakha Sabharwal
- Division of Pediatrics Infectious Disease, Department of Pediatrics, Boston Medical Center, Boston, MA, US.
| |
Collapse
|
28
|
Hanke M, Dijkstra L, Foraita R, Didelez V. Variable selection in linear regression models: Choosing the best subset is not always the best choice. Biom J 2024; 66:e2200209. [PMID: 37643390 DOI: 10.1002/bimj.202200209] [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] [Received: 07/31/2022] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 08/31/2023]
Abstract
We consider the question of variable selection in linear regressions, in the sense of identifying the correct direct predictors (those variables that have nonzero coefficients given all candidate predictors). Best subset selection (BSS) is often considered the "gold standard," with its use being restricted only by its NP-hard nature. Alternatives such as the least absolute shrinkage and selection operator (Lasso) or the Elastic net (Enet) have become methods of choice in high-dimensional settings. A recent proposal represents BSS as a mixed-integer optimization problem so that large problems have become computationally feasible. We present an extensive neutral comparison assessing the ability to select the correct direct predictors of BSS compared to forward stepwise selection (FSS), Lasso, and Enet. The simulation considers a range of settings that are challenging regarding dimensionality (number of observations and variables), signal-to-noise ratios, and correlations between predictors. As fair measure of performance, we primarily used the best possible F1-score for each method, and results were confirmed by alternative performance measures and practical criteria for choosing the tuning parameters and subset sizes. Surprisingly, it was only in settings where the signal-to-noise ratio was high and the variables were uncorrelated that BSS reliably outperformed the other methods, even in low-dimensional settings. Furthermore, FSS performed almost identically to BSS. Our results shed new light on the usual presumption of BSS being, in principle, the best choice for selecting the correct direct predictors. Especially for correlated variables, alternatives like Enet are faster and appear to perform better in practical settings.
Collapse
Affiliation(s)
- Moritz Hanke
- Department of Biometry and Data Management, Leibniz Institute for Prevention Research and Epidemiology - BIPS, Bremen, Germany
| | - Louis Dijkstra
- Department of Biometry and Data Management, Leibniz Institute for Prevention Research and Epidemiology - BIPS, Bremen, Germany
| | - Ronja Foraita
- Department of Biometry and Data Management, Leibniz Institute for Prevention Research and Epidemiology - BIPS, Bremen, Germany
| | - Vanessa Didelez
- Department of Biometry and Data Management, Leibniz Institute for Prevention Research and Epidemiology - BIPS, Bremen, Germany
- Department of Mathematics and Computer Science, University of Bremen, Bremen, Germany
| |
Collapse
|
29
|
Gao C, Chen Q, Hao X, Wang Q. Immunomodulation of Antibody Glycosylation through the Placental Transfer. Int J Mol Sci 2023; 24:16772. [PMID: 38069094 PMCID: PMC10705935 DOI: 10.3390/ijms242316772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Establishing an immune balance between the mother and fetus during gestation is crucial, with the placenta acting as the epicenter of immune tolerance. The placental transfer of antibodies, mainly immunoglobulin G (IgG), is critical in protecting the developing fetus from infections. This review looks at how immunomodulation of antibody glycosylation occurs during placental transfer and how it affects fetal health. The passage of maternal IgG antibodies through the placental layers, including the syncytiotrophoblast, stroma, and fetal endothelium, is discussed. The effect of IgG subclass, glycosylation, concentration, maternal infections, and antigen specificity on antibody transfer efficiency is investigated. FcRn-mediated IgG transport, influenced by pH-dependent binding, is essential for placental transfer. Additionally, this review delves into the impact of glycosylation patterns on antibody functionality, considering both protective and pathological effects. Factors affecting the transfer of protective antibodies, such as maternal vaccination, are discussed along with reducing harmful antibodies. This in-depth examination of placental antibody transfer and glycosylation provides insights into improving neonatal immunity and mitigating the effects of maternal autoimmune and alloimmune conditions.
Collapse
Affiliation(s)
| | | | | | - Qiushi Wang
- Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
30
|
Keitany GJ, Rubin BER, Garrett ME, Musa A, Tracy J, Liang Y, Ebert P, Moore AJ, Guan J, Eggers E, Lescano N, Brown R, Carbo A, Al-Asadi H, Ching T, Day A, Harris R, Linkem C, Popov D, Wilkins C, Li L, Wang J, Liu C, Chen L, Dines JN, Atyeo C, Alter G, Baldo L, Sherwood A, Howie B, Klinger M, Yusko E, Robins HS, Benzeno S, Gilbert AE. Multimodal, broadly neutralizing antibodies against SARS-CoV-2 identified by high-throughput native pairing of BCRs from bulk B cells. Cell Chem Biol 2023; 30:1377-1389.e8. [PMID: 37586370 PMCID: PMC10659930 DOI: 10.1016/j.chembiol.2023.07.011] [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] [Received: 09/20/2022] [Revised: 04/25/2023] [Accepted: 07/23/2023] [Indexed: 08/18/2023]
Abstract
TruAB Discovery is an approach that integrates cellular immunology, high-throughput immunosequencing, bioinformatics, and computational biology in order to discover naturally occurring human antibodies for prophylactic or therapeutic use. We adapted our previously described pairSEQ technology to pair B cell receptor heavy and light chains of SARS-CoV-2 spike protein-binding antibodies derived from enriched antigen-specific memory B cells and bulk antibody-secreting cells. We identified approximately 60,000 productive, in-frame, paired antibody sequences, from which 2,093 antibodies were selected for functional evaluation based on abundance, isotype and patterns of somatic hypermutation. The exceptionally diverse antibodies included RBD-binders with broad neutralizing activity against SARS-CoV-2 variants, and S2-binders with broad specificity against betacoronaviruses and the ability to block membrane fusion. A subset of these RBD- and S2-binding antibodies demonstrated robust protection against challenge in hamster and mouse models. This high-throughput approach can accelerate discovery of diverse, multifunctional antibodies against any target of interest.
Collapse
Affiliation(s)
| | | | | | - Andrea Musa
- Adaptive Biotechnologies, Seattle, WA 98109, USA
| | - Jeff Tracy
- Adaptive Biotechnologies, Seattle, WA 98109, USA
| | - Yu Liang
- Adaptive Biotechnologies, Seattle, WA 98109, USA
| | - Peter Ebert
- Adaptive Biotechnologies, Seattle, WA 98109, USA
| | | | | | - Erica Eggers
- Adaptive Biotechnologies, Seattle, WA 98109, USA
| | | | - Ryan Brown
- Adaptive Biotechnologies, Seattle, WA 98109, USA
| | - Adria Carbo
- Adaptive Biotechnologies, Seattle, WA 98109, USA
| | | | | | - Austin Day
- Adaptive Biotechnologies, Seattle, WA 98109, USA
| | | | | | | | | | - Lianqu Li
- GenScript ProBio Biotech, Nanjing, Jiangsu Province, China
| | - Jiao Wang
- GenScript ProBio Biotech, Nanjing, Jiangsu Province, China
| | - Chuanxin Liu
- GenScript ProBio Biotech, Nanjing, Jiangsu Province, China
| | - Li Chen
- GenScript ProBio Biotech, Nanjing, Jiangsu Province, China
| | | | - Caroline Atyeo
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Lance Baldo
- Adaptive Biotechnologies, Seattle, WA 98109, USA
| | | | - Bryan Howie
- Adaptive Biotechnologies, Seattle, WA 98109, USA
| | - Mark Klinger
- Adaptive Biotechnologies, Seattle, WA 98109, USA
| | - Erik Yusko
- Adaptive Biotechnologies, Seattle, WA 98109, USA
| | | | | | | |
Collapse
|
31
|
Wessel RE, Dolatshahi S. Quantitative mechanistic model reveals key determinants of placental IgG transfer and informs prenatal immunization strategies. PLoS Comput Biol 2023; 19:e1011109. [PMID: 37934786 PMCID: PMC10656024 DOI: 10.1371/journal.pcbi.1011109] [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: 04/17/2023] [Revised: 11/17/2023] [Accepted: 10/12/2023] [Indexed: 11/09/2023] Open
Abstract
Transplacental antibody transfer is crucially important in shaping neonatal immunity. Recently, prenatal maternal immunization has been employed to boost pathogen-specific immunoglobulin G (IgG) transfer to the fetus. Multiple factors have been implicated in antibody transfer, but how these key regulators work together to elicit selective transfer is pertinent to engineering vaccines for mothers to optimally immunize their newborns. Here, we present the first quantitative mechanistic model to uncover the determinants of placental antibody transfer and inform personalized immunization approaches. We identified placental FcγRIIb expressed by endothelial cells as a limiting factor in receptor-mediated transfer, which plays a key role in promoting preferential transport of subclasses IgG1, IgG3, and IgG4, but not IgG2. Integrated computational modeling and in vitro experiments reveal that IgG subclass abundance, Fc receptor (FcR) binding affinity, and FcR abundance in syncytiotrophoblasts and endothelial cells contribute to inter-subclass competition and potentially inter- and intra-patient antibody transfer heterogeneity. We developed an in silico prenatal vaccine testbed by combining a computational model of maternal vaccination with this placental transfer model using the tetanus, diphtheria, and acellular pertussis (Tdap) vaccine as a case study. Model simulations unveiled precision prenatal immunization opportunities that account for a patient's anticipated gestational length, placental size, and FcR expression by modulating vaccine timing, dosage, and adjuvant. This computational approach provides new perspectives on the dynamics of maternal-fetal antibody transfer in humans and potential avenues to optimize prenatal vaccinations that promote neonatal immunity.
Collapse
Affiliation(s)
- Remziye E. Wessel
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Sepideh Dolatshahi
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| |
Collapse
|
32
|
Abdolahi T, Maamouri G, Behmadi M, Mirzaeian S, Boskabadi H, Faramarzi R. Investigating the impact of Sinopharm COVID-19 vaccination on antibody response in pregnant women and their newborns. J Med Virol 2023; 95:e29231. [PMID: 37971780 DOI: 10.1002/jmv.29231] [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: 06/19/2023] [Revised: 10/22/2023] [Accepted: 10/31/2023] [Indexed: 11/19/2023]
Abstract
This study aims to investigate the levels of receptor-binding domain (RBD), spike, and neutralizing antibodies in pregnant women who received the Sinopharm vaccine and their newborns. A cross-sectional study was conducted at a tertiary center, Mashhad, Iran. We included 88 pregnant women who had received at least two doses of the Sinopharm vaccine. Maternal and umbilical cord blood samples taken at delivery were analyzed for antibodies using ELISA tests. Antibody levels did not vary significantly between women with two or three vaccine doses. Only 1.1% of mothers had undetectable levels of RBD antibodies, but detectable antibodies were observed in all newborns. A significant linear correlation was found between the levels of neutralizing antibodies (r = 0.7, p < 0.001) and RBD antibodies (r = 0.833, p < 0.001) in mothers and their newborns, but not for Spike antibodies (r = 0.214, p = 0.045). In mothers, high titers of antispike and RBD antibodies were observed at the time of delivery. The high titers of RBD and antispike antibodies were found in cord blood, suggesting potential neonatal immunity. Detectable levels of antibodies were found in both groups, regardless of the timing of vaccination. The Sinopharm vaccine generates detectable levels of antibodies in pregnant women, which are efficiently transferred to their newborns. The number of vaccine doses (two or three) did not significantly impact the levels of detectable antibodies. This underscores Sinopharm's potential efficacy in protecting pregnant women and their infants from COVID-19.
Collapse
Affiliation(s)
- Tahoora Abdolahi
- Neonatal Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamali Maamouri
- Department of Pediatrics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Behmadi
- Department of Pediatrics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sara Mirzaeian
- Department of obstetrics and gynecology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hassan Boskabadi
- Department of Pediatrics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Raheleh Faramarzi
- Department of Pediatrics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
33
|
Spatola M, Nziza N, Jung W, Deng Y, Yuan D, Dinoto A, Bozzetti S, Chiodega V, Ferrari S, Lauffenburger DA, Mariotto S, Alter G. Neurologic sequelae of COVID-19 are determined by immunologic imprinting from previous coronaviruses. Brain 2023; 146:4292-4305. [PMID: 37161609 PMCID: PMC11004923 DOI: 10.1093/brain/awad155] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/28/2023] [Accepted: 04/10/2023] [Indexed: 05/11/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), which is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), remains a global public health emergency. Although SARS-CoV-2 is primarily a respiratory pathogen, extra-respiratory organs, including the CNS, can also be affected. Neurologic symptoms have been observed not only during acute SARS-CoV-2 infection, but also at distance from respiratory disease, also known as long-COVID or neurological post-acute sequelae of COVID-19 (neuroPASC). The pathogenesis of neuroPASC is not well understood, but hypotheses include SARS-CoV-2-induced immune dysfunctions, hormonal dysregulations and persistence of SARS-CoV-2 reservoirs. In this prospective cohort study, we used a high throughput systems serology approach to dissect the humoral response to SARS-CoV-2 (and other common coronaviruses: 229E, HKU1, NL63 and OC43) in the serum and CSF from 112 infected individuals who developed (n = 18) or did not develop (n = 94) neuroPASC. Unique SARS-CoV-2 humoral profiles were observed in the CSF of neuroPASC compared with serum responses. All antibody isotypes (IgG, IgM, IgA) and subclasses (IgA1-2, IgG1-4) were detected in serum, whereas CSF was characterized by focused IgG1 (and absence of IgM). These data argue in favour of compartmentalized brain-specific responses against SARS-CoV-2 through selective transfer of antibodies from the serum to the CSF across the blood-brain barrier, rather than intrathecal synthesis, where more diversity in antibody classes/subclasses would be expected. Compared to individuals who did not develop post-acute complications following infection, individuals with neuroPASC had similar demographic features (median age 65 versus 66.5 years, respectively, P = 0.55; females 33% versus 44%, P = 0.52) but exhibited attenuated systemic antibody responses against SARS-CoV-2, characterized by decreased capacity to activate antibody-dependent complement deposition (ADCD), NK cell activation (ADNKA) and to bind Fcγ receptors. However, surprisingly, neuroPASC individuals showed significantly expanded antibody responses to other common coronaviruses, including 229E, HKU1, NL63 and OC43. This biased humoral activation across coronaviruses was particularly enriched in neuroPASC individuals with poor outcome, suggesting an 'original antigenic sin' (or immunologic imprinting), where pre-existing immune responses against related viruses shape the response to the current infection, as a key prognostic marker of neuroPASC disease. Overall, these findings point to a pathogenic role for compromised anti-SARS-CoV-2 responses in the CSF, likely resulting in incomplete virus clearance from the brain and persistent neuroinflammation, in the development of post-acute neurologic complications of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Marianna Spatola
- Ragon Institute of MGH, MIT and Harvard Medical School, Cambridge, MA 02139, USA
| | - Nadège Nziza
- Ragon Institute of MGH, MIT and Harvard Medical School, Cambridge, MA 02139, USA
| | - Wonyeong Jung
- Ragon Institute of MGH, MIT and Harvard Medical School, Cambridge, MA 02139, USA
- Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yixiang Deng
- Ragon Institute of MGH, MIT and Harvard Medical School, Cambridge, MA 02139, USA
- Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dansu Yuan
- Ragon Institute of MGH, MIT and Harvard Medical School, Cambridge, MA 02139, USA
| | - Alessandro Dinoto
- Neurology Unit, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37131 Verona, Italy
| | - Silvia Bozzetti
- Neurology Unit, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37131 Verona, Italy
| | - Vanessa Chiodega
- Neurology Unit, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37131 Verona, Italy
- Department of Neurology/Stroke Unit, San Maurizio Hospital, 39100 Bolzano, Italy
| | - Sergio Ferrari
- Neurology Unit, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37131 Verona, Italy
| | | | - Sara Mariotto
- Neurology Unit, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37131 Verona, Italy
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard Medical School, Cambridge, MA 02139, USA
| |
Collapse
|
34
|
Liu S, Wang P, Shi X, Weng T, Zhong J, Zhang X, Qu J, Chen L, Xu Q, Meng X, Xiong H, Wu D, Fang D, Peng B, Zhang D. Maternal antibody transfer rate of vaccination against SARS-CoV-2 before or during early pregnancy and its protective effectiveness on offspring. J Med Virol 2023; 95:e29125. [PMID: 37800607 DOI: 10.1002/jmv.29125] [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: 07/22/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 10/07/2023]
Abstract
This study focuses on maternal antibody transfer following vaccination against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) before or during early pregnancy and its potential protective effects on infants, providing scientific evidence for vaccination strategies. This prospective study tested the samples for SARS-CoV-2 IgG antibody titers and neutralizing capacity and tracked the infections after birth. Perform multivariate analysis of factors influencing antibody transfer rate, newborn antibody titers, and infant infection. Total 87.1% (122/140) women received coronavirus disease 2019 (COVID-19) vaccine before or during early pregnancy, and 28 of them had breakthrough infection. The maternal and neonatal IgG positive rates at delivery were 60.7% (85/140) and 60.8% (87/143), respectively. A positive correlation was found between neonatal and maternal IgG antibody titers. Compared with the median IgG antibody transfer rate of infected pregnant women, that of vaccinated but not infected pregnant women was higher (1.21 versus: 1.53 [two doses], 1.71 [three doses]). However, neonatal IgG antibodies were relatively low (174.91 versus: 0.99 [two doses], 8.18 [three doses]), and their neutralizing capacity was weak. The overall effectiveness of maternal vaccination in preventing infant infection was 27.0%, and three doses had higher effectiveness than two doses (64.3% vs. 19.6%). Multivariate analysises showed that in vaccination group women receiving three doses or in infection group women with longer interval between infection and delivery had a higher antibody transfer rate and neonatal IgG antibody titer. More than half of women vaccinated before or during early pregnancy can achieve effective antibody transfer to newborns. However, the neonatal IgG antibody titer is low and has a weak neutralizing capacity, providing limited protection to infants.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Ping Wang
- Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, China
| | - Xiaolu Shi
- Department of Microbiology Laboratory, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Tingsong Weng
- Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, China
| | - Jiayi Zhong
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Xiaomin Zhang
- Department of Microbiology Laboratory, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Jing Qu
- Department of Microbiology Laboratory, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Long Chen
- Department of Microbiology Laboratory, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Qing Xu
- Department of Microbiology Laboratory, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Xiang Meng
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Husheng Xiong
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Dawei Wu
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Dajun Fang
- Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, China
| | - Bo Peng
- Department of Microbiology Laboratory, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Dingmei Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
35
|
Carrasco Colom J, Manzanares Á, Álvaro Gómez A, Serrano Escribano I, Esquivel E, Pérez-Rivilla A, Moral-Pumarega MT, Aguirre Pascual E, De Vergas J, Reda Del Barrio S, Moraleda C, Epalza C, Fernández-Cooke E, Prieto L, Villaverde S, Zamora B, Herraiz I, Galindo A, Folgueira MD, Delgado R, Blázquez-Gamero D. Clinical outcomes and antibody transfer in a cohort of infants with in utero or perinatal exposure to SARS-CoV-2 (Coronascope Study). Eur J Pediatr 2023; 182:4647-4654. [PMID: 37561198 DOI: 10.1007/s00431-023-05147-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023]
Abstract
We aimed to describe the outcomes, focusing on the hearing and neurological development, of infants born to mothers with COVID-19 during pregnancy and to evaluate the persistence of maternal antibodies in the first months of life. An observational, prospective study at a tertiary hospital in Madrid (Spain) on infants born to mothers with COVID-19 during pregnancy between March and September 2020 was conducted. A follow-up visit at 1-3 months of age with a physical and neurological examination, cranial ultrasound (cUS), SARS-CoV-2 RT-PCR on nasopharyngeal swab, and SARS-CoV-2 serology were performed. Hearing was evaluated at birth through the automated auditory brainstem response and at six months of age through the auditory steady-state response. A neurodevelopmental examination using the Bayley-III scale was performed at 12 months of age. Of 95 infants studied, neurological examination was normal in all of them at the follow-up visit, as was the cUS in 81/85 (95%) infants, with only mild abnormalities in four of them. Serology was positive in 47/95 (50%) infants, which was not associated with symptoms or severity of maternal infection. No hearing loss was detected, and neurodevelopment was normal in 96% of the infants (median Z score: 0). CONCLUSION In this cohort, the majority of infants born to mothers with COVID-19 during pregnancy were healthy infants with a normal cUS, no hearing loss, and normal neurodevelopment in the first year of life. Only half of the infants had a positive serological result during the follow-up. WHAT IS KNOWN • Hearing loss and neurodevelopmental delay in infants born to mothers with COVID-19 during pregnancy has been suggested, although data is inconsistent. Maternal antibody transfer seems to be high, with a rapid decrease during the first weeks of life. WHAT IS NEW • Most infants born to mothers with COVID-19 during pregnancy had normal hearing screening, cranial ultrasound, and neurodevelopmental status at 12 months of life. Antibodies against SARS-CoV-2 were only detected in 50% of the infants at two months of life.
Collapse
Affiliation(s)
- Jaime Carrasco Colom
- Pediatric Infectious Disease Unit, Hospital Universitario 12 de Octubre, Madrid, Spain
- Pediatric Research and Clinical Trials Unit (UPIC), Instituto de Investigación Hospital, RECLIP, 12 de Octubre (imas12), Madrid, Spain
| | - Ángela Manzanares
- Pediatric Infectious Disease Unit, Hospital Universitario 12 de Octubre, Madrid, Spain.
- Pediatric Research and Clinical Trials Unit (UPIC), Instituto de Investigación Hospital, RECLIP, 12 de Octubre (imas12), Madrid, Spain.
| | | | | | - Estrella Esquivel
- Pediatric Research and Clinical Trials Unit (UPIC), Instituto de Investigación Hospital, RECLIP, 12 de Octubre (imas12), Madrid, Spain
| | - Alfredo Pérez-Rivilla
- Microbiology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Maria Teresa Moral-Pumarega
- Neonatology Department, Hospital Universitario 12 de Octubre, RICORS Network, ISCIII, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
| | | | - Joaquín De Vergas
- Pediatric Otorhinolaryngology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Sara Reda Del Barrio
- Pediatric Otorhinolaryngology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Cinta Moraleda
- Pediatric Infectious Disease Unit, Hospital Universitario 12 de Octubre, Madrid, Spain
- Pediatric Research and Clinical Trials Unit (UPIC), Instituto de Investigación Hospital, RECLIP, 12 de Octubre (imas12), Madrid, Spain
| | - Cristina Epalza
- Pediatric Infectious Disease Unit, Hospital Universitario 12 de Octubre, Madrid, Spain
- Pediatric Research and Clinical Trials Unit (UPIC), Instituto de Investigación Hospital, RECLIP, 12 de Octubre (imas12), Madrid, Spain
| | - Elisa Fernández-Cooke
- Pediatric Infectious Disease Unit, Hospital Universitario 12 de Octubre, Madrid, Spain
- Pediatric Research and Clinical Trials Unit (UPIC), Instituto de Investigación Hospital, RECLIP, 12 de Octubre (imas12), Madrid, Spain
| | - Luis Prieto
- Pediatric Infectious Disease Unit, Hospital Universitario 12 de Octubre, Madrid, Spain
- Pediatric Research and Clinical Trials Unit (UPIC), Instituto de Investigación Hospital, RECLIP, 12 de Octubre (imas12), Madrid, Spain
- Universidad Complutense de Madrid, Madrid, Spain
| | - Serena Villaverde
- Pediatric Infectious Disease Unit, Hospital Universitario 12 de Octubre, Madrid, Spain
- Pediatric Research and Clinical Trials Unit (UPIC), Instituto de Investigación Hospital, RECLIP, 12 de Octubre (imas12), Madrid, Spain
| | - Berta Zamora
- Neuropsychology Unit, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Ignacio Herraiz
- Universidad Complutense de Madrid, Madrid, Spain
- Fetal Medicine Unit, Obstetrics and Gynecology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
- Primary Care Interventions to Prevent Chronic Maternal and Child Diseases of Perinatal and Developmental Origin (RICORS Network), Instituto de Salud Carlos III, RD21/0012/0024, Madrid, Spain
| | - Alberto Galindo
- Universidad Complutense de Madrid, Madrid, Spain
- Fetal Medicine Unit, Obstetrics and Gynecology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
- Primary Care Interventions to Prevent Chronic Maternal and Child Diseases of Perinatal and Developmental Origin (RICORS Network), Instituto de Salud Carlos III, RD21/0012/0024, Madrid, Spain
| | - María Dolores Folgueira
- Microbiology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Rafael Delgado
- Microbiology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Daniel Blázquez-Gamero
- Pediatric Infectious Disease Unit, Hospital Universitario 12 de Octubre, Madrid, Spain
- Pediatric Research and Clinical Trials Unit (UPIC), Instituto de Investigación Hospital, RECLIP, 12 de Octubre (imas12), Madrid, Spain
- Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
36
|
Mahant AM, Trejo FE, Aguilan JT, Sidoli S, Permar SR, Herold BC. Antibody attributes, Fc receptor expression, gestation and maternal SARS-CoV-2 infection modulate HSV IgG placental transfer. iScience 2023; 26:107648. [PMID: 37670782 PMCID: PMC10475509 DOI: 10.1016/j.isci.2023.107648] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/30/2023] [Accepted: 08/11/2023] [Indexed: 09/07/2023] Open
Abstract
Antibody-dependent cellular cytotoxicity (ADCC) is associated with protection against neonatal herpes. We hypothesized that placental transfer of ADCC-mediating herpes simplex virus (HSV) immunoglobulin G (IgG) is influenced by antigenic target, function, glycans, gestational age, and maternal severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Maternal and cord blood were collected from HSV-seropositive (HSV+) mothers pre-COVID and HSV+/SARS-CoV-2+ mothers during the pandemic. Transfer of HSV neutralizing IgG was significantly lower in preterm versus term dyads (transfer ratio [TR] 0.84 vs. 2.44) whereas the TR of ADCC-mediating IgG was <1.0 in both term and preterm pre-COVID dyads. Anti-glycoprotein D IgG, which had only neutralizing activity, and anti-glycoprotein B (gB) IgG, which displayed neutralizing and ADCC activity, exhibited different relative affinities for the neonatal Fc receptor (FcRn) and expressed different glycans. The transfer of ADCC-mediating IgG increased significantly in term SARS-CoV-2+ dyads. This was associated with greater placental colocalization of FcRn with FcγRIIIa. These findings have implications for strategies to prevent neonatal herpes.
Collapse
Affiliation(s)
- Aakash Mahant Mahant
- Departments of Microbiology and Immunology, Obstetrics-Gynecology and Women’s Health, and Biochemistry Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Fatima Estrada Trejo
- Departments of Microbiology and Immunology, Obstetrics-Gynecology and Women’s Health, and Biochemistry Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jennifer T. Aguilan
- Departments of Microbiology and Immunology, Obstetrics-Gynecology and Women’s Health, and Biochemistry Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Simone Sidoli
- Departments of Microbiology and Immunology, Obstetrics-Gynecology and Women’s Health, and Biochemistry Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Sallie R. Permar
- Department of Pediatrics, Weil Cornell Medicine, New York, NY 10021, USA
| | - Betsy C. Herold
- Departments of Microbiology and Immunology, Obstetrics-Gynecology and Women’s Health, and Biochemistry Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
37
|
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
|
38
|
Habbash AS, Siddiqui AF. Factors Affecting COVID-19 Vaccine Acceptance among Pregnant Women: A Cross Sectional Study from Abha City, Saudi Arabia. Vaccines (Basel) 2023; 11:1463. [PMID: 37766139 PMCID: PMC10536313 DOI: 10.3390/vaccines11091463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Background: Pregnant women can get infected with COVID-19 with serious sequelae to them and their fetus. Concerns about COVID-19 vaccination safety to mothers and babies, and doubts about its effectiveness, have hindered vaccine acceptance throughout the COVID-19 crisis. The objective of the current investigation was to estimate COVID-19 acceptance rates among pregnant women in Abha city, Aseer region, Saudi Arabia, and determine its clinical and demographic correlates. Method: Descriptive questionnaire-based cross-sectional survey of a sample of pregnant women attending regular antenatal care services in Abha. We used backward stepwise multiple logistic regression analysis to evaluate the predictability of vaccine acceptance in terms of baseline clinical and demographic factors. Results: The survey included 572 pregnant women. The prevalence of acceptance of COVID-19 vaccine was high (93.7%; 95%CI: 91.7-95.7%). University graduates and women with a later gestational age were more likely to accept vaccination (OR = 6.120, p = 0.009), (t = 2.163, p = 0.036), respectively. Confidence in vaccine safety was associated with better acceptance (OR = 3.431, p = 0.001). Conclusions: The acceptance rate for vaccination among pregnant women in Abha, Saudi Arabia, is higher compared to international rate. However, our results indicate that confidence in vaccine safety was associated with better acceptance. Hence, vaccine safety was the overarching predictor for harboring positive attitudes towards it. Public health policies should capitalize on such positive attitudes and aim for total coverage of pregnant women with COVID-19 vaccination including booster dosages.
Collapse
Affiliation(s)
| | - Aesha Farheen Siddiqui
- Department of Family and Community Medicine, College of Medicine, King Khalid University, Abha 62529, Saudi Arabia
| |
Collapse
|
39
|
Vigil-Vázquez S, Manzanares Á, Hernanz-Lobo A, Carrasco-García I, Zamora Del Pozo C, Pérez-Pérez A, Rincón-López EM, Santiago-García B, Pintado-Recarte MDP, Alonso-Fernández R, Sánchez-Luna M, Navarro-Gómez ML. Serologic evolution and follow-up to IgG antibodies of infants born to mothers with gestational COVID. BMC Pregnancy Childbirth 2023; 23:623. [PMID: 37648971 PMCID: PMC10469412 DOI: 10.1186/s12884-023-05926-6] [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] [Received: 02/25/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND It is known that SARS-CoV-2 antibodies from pregnant women with SARS-CoV-2 infection during pregnancy cross the placenta but the duration and the protective effect of these antibodies in infants is scarce. METHODS This prospective study included mothers with SARS-COV-2 infection during pregnancy and their infants from April 2020 to March 2021. IgG antibodies to SARS-CoV-2 spike protein were performed on women and infants at birth and at two and six months during follow-up. Anthropometrical measures and physical and neurological examinations and a clinical history of symptoms and COVID-19 diagnosis were collected. Simple linear regression was performed to compare categorical and continuous variables. To compare the mother's and infant's antibody titers evolution, a mixed linear regression model was used. A predictive model of newborn antibody titers at birth has been established by means of simple stepwise linear regression. RESULTS 51 mother-infant couples were included. 45 (90%) of the mothers and 44 (86.3%) of the newborns had a positive serology al birth. These antibodies were progressively decreasing and were positive in 34 (66.7%) and 7 (13.7%) of infants at 2 and 6 months, respectively. IgG titers of newborns at birth were related to mothers' titers, with a positive moderate correlation (Pearson's correlation coefficient: 0.82, p < 0,001). Fetal/maternal antibodies placental transference rate was 1.3 (IQR: 0.7-2.2). The maternal IgG titers at delivery and the type of maternal infection (acute, recent, or past infection) was significantly related with infants' antibody titers at birth. No other epidemiological or clinical factors were related to antibodies titers. Neurodevelopment, psychomotor development, and growth were normal in 94.2% of infants in the third follow-up visit. No infants had a COVID-19 diagnosis during the follow-up period. CONCLUSIONS Transplacental transfer of maternal antibodies is high in newborns from mothers with recent or past infection at delivery, but these antibodies decrease after the first months of life. Infant's IgG titers were related to maternal IgG titers at delivery. Further studies are needed to learn about the protective role of maternal antibodies in infants.
Collapse
Affiliation(s)
- Sara Vigil-Vázquez
- Department of Neonatology, Hospital General Universitario Gregorio Marañón, Calle O'Donnel 48, Madrid, 28009, Spain.
| | - Ángela Manzanares
- Pediatric Infectious Disease Unit, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Pediatrics Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Alicia Hernanz-Lobo
- Pediatric Infectious Disease Unit, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Pediatrics Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Itziar Carrasco-García
- Pediatric Infectious Disease Unit, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Pediatrics Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Clara Zamora Del Pozo
- Department of Obstetrics and Gynecology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Alba Pérez-Pérez
- Department of Neonatology, Hospital General Universitario Gregorio Marañón, Calle O'Donnel 48, Madrid, 28009, Spain
| | - Elena María Rincón-López
- Pediatric Infectious Disease Unit, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Pediatrics Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Begoña Santiago-García
- Pediatric Infectious Disease Unit, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Pediatrics Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Roberto Alonso-Fernández
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Manuel Sánchez-Luna
- Department of Neonatology, Hospital General Universitario Gregorio Marañón, Calle O'Donnel 48, Madrid, 28009, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - María Luisa Navarro-Gómez
- Pediatric Infectious Disease Unit, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Pediatrics Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
40
|
Brebant D, Couffignal C, Manchon P, Duquesne S, Picone O, Vauloup-Fellous C. Transplacental transfer of anti-SARS-CoV-2 neutralizing antibodies in comparison to other pathogens total antibodies. J Clin Virol 2023; 165:105495. [PMID: 37295035 PMCID: PMC10212596 DOI: 10.1016/j.jcv.2023.105495] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/16/2023] [Accepted: 05/22/2023] [Indexed: 06/11/2023]
Abstract
BACKGROUNDS Due to immaturity of their immune system, passive maternal immunization is essential for newborns during their first months of life. Therefore, in the current context of intense circulation of SARS-CoV-2, identifying factors influencing the transfer ratio (TR) of neutralizing antibodies against SARS-CoV-2 (NAb) appears important. METHODS Our study nested in the COVIPREG cohort (NCT04355234), included mothers who had a SARS-CoV-2 PCR positive during their pregnancy and their newborns. Maternal and neonatal NAb levels were measured with the automated iFlash system. RESULTS For the 173 mother-infant pairs included in our study, the median gestational age (GA) at delivery was 39.4 weeks of gestation (WG), and 29.7 WG at maternal SARS-CoV-2 infection. Using a multivariate logistic model, having a NAb TR above 1 was positively associated with a longer delay from maternal positive SARS-CoV-2 PCR to delivery (aOR 1.09, 95% CI: 1.03 - 1.17) and with a later GA at delivery (aOR = 1.58, 95% CI: 1.09 - 2.52). It was negatively associated with being a male newborn (aOR 0.21, 95% CI: 0.07 - 0.59). In 3rd trimester SARS-CoV-2 infected mothers, NAb TR was inferior to VZV, toxoplasmosis, CMV, measle and rubella's TR. However, in 1st or 2nd trimester infected mothers, only measle TR was different from NAb TR. CONCLUSION Male newborn of mothers infected by SARS-CoV-2 during their pregnancy appear to have less protection against SARS-CoV-2 in their first months of life than female newborns. Measle TR was superior to NAb TR even in case of 1st or 2nd trimester maternal SARS-CoV-2 infection. Future studies are needed to investigate possible differences in transmission of NAb following infection vs vaccination and its impact on TR.
Collapse
MESH Headings
- SARS-CoV-2/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Maternal-Fetal Exchange/immunology
- Gestational Age
- Humans
- Male
- Female
- COVID-19/blood
- COVID-19/immunology
- COVID-19/prevention & control
- Delivery, Obstetric
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Pregnancy
- Infant, Newborn
- Sex Characteristics
- COVID-19 Vaccines
- Vaccination
- Pregnancy Complications/blood
- Pregnancy Complications/immunology
- Infant, Newborn, Diseases/immunology
- Infant, Newborn, Diseases/prevention & control
- Infectious Disease Transmission, Vertical/prevention & control
- Paris
- Adult
Collapse
Affiliation(s)
- Diane Brebant
- Université Paris-Saclay, Inserm U1193, AP-HP, Hôpital Paul Brousse, Virology department, Villejuif 94800, France; Groupe de Recherche sur les Infections pendant la Grossesse (GRIG), France.
| | - Camille Couffignal
- Université Paris Cité, INSERM CIC-EC 1425, AP-HP, Hôpital Bichat - Claude Bernard, Clinical Research Department, France
| | - Pauline Manchon
- Université Paris Cité, INSERM CIC-EC 1425, AP-HP, Hôpital Bichat - Claude Bernard, Clinical Research Department, France
| | - Sandra Duquesne
- Université Paris-Saclay, Inserm U1193, AP-HP, Hôpital Paul Brousse, Virology department, Villejuif 94800, France
| | - Olivier Picone
- Université Paris Cité, Hôpital louis Mourier, Obstetrical department, France; IAME U1137, Inserm, Université Paris Cité, France; Groupe de Recherche sur les Infections pendant la Grossesse (GRIG), France
| | - Christelle Vauloup-Fellous
- Université Paris-Saclay, Inserm U1193, AP-HP, Hôpital Paul Brousse, Virology department, Villejuif 94800, France; Groupe de Recherche sur les Infections pendant la Grossesse (GRIG), France.
| |
Collapse
|
41
|
Wijenayake S, Martz J, Lapp HE, Storm JA, Champagne FA, Kentner AC. The contributions of parental lactation on offspring development: It's not udder nonsense! Horm Behav 2023; 153:105375. [PMID: 37269591 PMCID: PMC10351876 DOI: 10.1016/j.yhbeh.2023.105375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 06/05/2023]
Abstract
The Developmental Origins of Health and Disease (DOHaD) hypothesis describes how maternal stress exposures experienced during critical periods of perinatal life are linked to altered developmental trajectories in offspring. Perinatal stress also induces changes in lactogenesis, milk volume, maternal care, and the nutritive and non-nutritive components of milk, affecting short and long-term developmental outcomes in offspring. For instance, selective early life stressors shape the contents of milk, including macro/micronutrients, immune components, microbiota, enzymes, hormones, milk-derived extracellular vesicles, and milk microRNAs. In this review, we highlight the contributions of parental lactation to offspring development by examining changes in the composition of breast milk in response to three well-characterized maternal stressors: nutritive stress, immune stress, and psychological stress. We discuss recent findings in human, animal, and in vitro models, their clinical relevance, study limitations, and potential therapeutic significance to improving human health and infant survival. We also discuss the benefits of enrichment methods and support tools that can be used to improve milk quality and volume as well as related developmental outcomes in offspring. Lastly, we use evidence-based primary literature to convey that even though select maternal stressors may modulate lactation biology (by influencing milk composition) depending on the severity and length of exposure, exclusive and/or prolonged milk feeding may attenuate the negative in utero effects of early life stressors and promote healthy developmental trajectories. Overall, scientific evidence supports lactation to be protective against nutritive and immune stressors, but the benefits of lactation in response to psychological stressors need further investigation.
Collapse
Affiliation(s)
- Sanoji Wijenayake
- Department of Biology, The University of Winnipeg, Winnipeg, Manitoba, Canada.
| | - Julia Martz
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA
| | - Hannah E Lapp
- Deparment of Psychology, University of Texas at Austin, Austin, TX, USA
| | - Jasmyne A Storm
- Department of Biology, The University of Winnipeg, Winnipeg, Manitoba, Canada
| | | | - Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA.
| |
Collapse
|
42
|
Purcell RA, Theisen RM, Arnold KB, Chung AW, Selva KJ. Polyfunctional antibodies: a path towards precision vaccines for vulnerable populations. Front Immunol 2023; 14:1183727. [PMID: 37600816 PMCID: PMC10433199 DOI: 10.3389/fimmu.2023.1183727] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/30/2023] [Indexed: 08/22/2023] Open
Abstract
Vaccine efficacy determined within the controlled environment of a clinical trial is usually substantially greater than real-world vaccine effectiveness. Typically, this results from reduced protection of immunologically vulnerable populations, such as children, elderly individuals and people with chronic comorbidities. Consequently, these high-risk groups are frequently recommended tailored immunisation schedules to boost responses. In addition, diverse groups of healthy adults may also be variably protected by the same vaccine regimen. Current population-based vaccination strategies that consider basic clinical parameters offer a glimpse into what may be achievable if more nuanced aspects of the immune response are considered in vaccine design. To date, vaccine development has been largely empirical. However, next-generation approaches require more rational strategies. We foresee a generation of precision vaccines that consider the mechanistic basis of vaccine response variations associated with both immunogenetic and baseline health differences. Recent efforts have highlighted the importance of balanced and diverse extra-neutralising antibody functions for vaccine-induced protection. However, in immunologically vulnerable populations, significant modulation of polyfunctional antibody responses that mediate both neutralisation and effector functions has been observed. Here, we review the current understanding of key genetic and inflammatory modulators of antibody polyfunctionality that affect vaccination outcomes and consider how this knowledge may be harnessed to tailor vaccine design for improved public health.
Collapse
Affiliation(s)
- Ruth A. Purcell
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Robert M. Theisen
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Kelly B. Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Amy W. Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Kevin J. Selva
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
43
|
Harakeh S, Khan IA, Rani GF, Ibrahim M, Khan AS, Almuhayawi M, Al-Raddadi R, Teklemariam AD, Hazzazi MS, Bawazir WM, Niyazi HA, Alamri T, Niyazi HA, Yousafzai YM. Transplacental Transfer of SARS-CoV-2 Receptor-Binding Domain IgG Antibodies from Mothers to Neonates in a Cohort of Pakistani Unvaccinated Mothers. Biomedicines 2023; 11:1651. [PMID: 37371746 DOI: 10.3390/biomedicines11061651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
The presence of COVID-19 antibodies in the maternal circulation is assumed to be protective for newborns against SARS-CoV-2 infection. We investigated whether maternal COVID-19 antibodies crossed the transplacental barrier and whether there was any difference in the hematological parameters of neonates born to mothers who recovered from COVID-19 during pregnancy. The cross-sectional study was conducted at the Saidu Group of Teaching Hospitals, located in Swat, Khyber Pakhtunkhwa. After obtaining written informed consent, 115 healthy, unvaccinated mother-neonate dyads were included. A clinical history of COVID-19-like illness, laboratory-confirmed diagnosis, and contact history were obtained. Serum samples from mothers and neonates were tested for SARS-CoV-2 anti-receptor-binding domain (anti-RBD) IgG antibodies. Hematological parameters were assessed with complete blood counts (CBC) and peripheral blood smear examinations. The study population consisted of 115 mothers, with a mean age of 29.44 ± 5.75 years, and most women (68/115 (59.1%)) were between 26 and 35 years of age. Of these mothers, 88/115 (76.5 percent) tested positive for SARS-CoV-2 anti-RBD IgG antibodies, as did 83/115 (72.2 percent) neonatal cord blood samples. The mean levels of SARS-CoV-2 IgG antibodies in maternal and neonatal blood were 19.86 ± 13.82 (IU/mL) and 16.16 ± 12.90 (IU/mL), respectively, indicating that maternal antibodies efficiently crossed the transplacental barrier with an antibody transfer ratio of 0.83. The study found no significant difference in complete blood count (CBC) parameters between seropositive and seronegative mothers, nor between neonates born to seropositive and seronegative mothers.
Collapse
Affiliation(s)
- Steve Harakeh
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Yousef Abdul Latif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah 22230, Saudi Arabia
| | - Ihsan Alam Khan
- Department of Hematology, Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar 25100, Pakistan
- Department of Pathology, Swat Medical College, Swat 19200, Pakistan
| | - Gulab Fatima Rani
- Department of Hematology, Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar 25100, Pakistan
| | - Muhammad Ibrahim
- Department of Hematology, Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar 25100, Pakistan
| | - Aysha Sarwar Khan
- Department of Hematology, Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar 25100, Pakistan
| | - Mohammed Almuhayawi
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Rajaa Al-Raddadi
- Community Medicine Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Addisu D Teklemariam
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohannad S Hazzazi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22254, Saudi Arabia
- Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 22254, Saudi Arabia
| | - Waleed M Bawazir
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22254, Saudi Arabia
- Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 22254, Saudi Arabia
| | - Hanouf A Niyazi
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Turki Alamri
- Family and Community Medicine Department, Faculty of Medicine in Rabigh, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hatoon A Niyazi
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Yasar Mehmood Yousafzai
- Department of Hematology, Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar 25100, Pakistan
- Rehman Medical Institute, Hayatabad Phase-V, Peshawar 25600, Pakistan
| |
Collapse
|
44
|
Martin‐Vicente M, Carrasco I, Muñoz‐Gomez M, Lobo AH, Mas V, Vigil‐Vázquez S, Vázquez M, Manzanares A, Cano O, Alonso R, Sepúlveda‐Crespo D, Tarancón‐Díez L, Muñoz‐Fernández M, Muñoz‐Chapuli M, Resino S, Navarro ML, Martinez I. Antibody levels to SARS-CoV-2 spike protein in mothers and children from delivery to six months later. Birth 2023; 50:418-427. [PMID: 35802776 PMCID: PMC9349436 DOI: 10.1111/birt.12667] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 05/11/2022] [Accepted: 06/15/2022] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Pregnant women are vulnerable to severe acute respiratory syndrome coronavirus (SARS-CoV-2) infection. Neutralizing antibodies against the SARS-CoV-2 spike (S) protein protect from severe disease. This study analyzes the antibody titers to SARS-CoV-2 S protein in pregnant women and their newborns at delivery, and six months later. METHODS We conducted a prospective study on pregnant women with confirmed SARS-CoV-2 infection and newborns. Antibody (IgG, IgM, and IgA) titers were determined using immunoassays in serum and milk samples. An angiotensin-converting enzyme 2 (ACE2) receptor-binding inhibition assay to the S protein was performed on the same serum and milk samples. RESULTS At birth, antibodies to SARS-CoV-2 spike protein were detected in 81.9% of mothers' sera, 78.9% of cord blood samples, and 63.2% of milk samples. Symptomatic women had higher antibody titers (IgG, IgM, and IgA) than the asymptomatic ones (P < 0.05). At six months postpartum, IgG levels decreased drastically in children's serum (P < 0.001) but remained high in mothers' serum. Antibody titers correlated positively with its capacity to inhibit the ACE2-spike protein interaction at baseline in maternal sera (R2 = 0.203; P < 0.001), cord sera (R2 = 0.378; P < 0.001), and milk (R2 = 0.564; P < 0.001), and at six months in maternal sera (R2 = 0.600; P < 0.001). CONCLUSIONS High antibody levels against SARS-CoV-2 spike protein were found in most pregnant women. Due to the efficient transfer of IgG to cord blood and high IgA titers in breast milk, neonates may be passively immunized to SARS-CoV-2 infection. Our findings could guide newborn management and maternal vaccination policies.
Collapse
Affiliation(s)
- María Martin‐Vicente
- Unidad de Infección Viral e InmunidadCentro Nacional de Microbiología, Instituto de Salud Carlos III, MajadahondaMadridSpain
| | - Itziar Carrasco
- Grupo de Investigación en Infectología PediátricaInstituto de Investigación Sanitaria Gregorio MarañónMadridSpain
| | - María José Muñoz‐Gomez
- Unidad de Infección Viral e InmunidadCentro Nacional de Microbiología, Instituto de Salud Carlos III, MajadahondaMadridSpain
| | - Alicia Hernanz Lobo
- Grupo de Investigación en Infectología PediátricaInstituto de Investigación Sanitaria Gregorio MarañónMadridSpain
- Servicio de PediatríaHospital General Universitario Gregorio MarañónMadridSpain
| | - Vicente Mas
- Unidad de Biología ViralCentro Nacional de Microbiología, Instituto de Salud Carlos IIIMadridSpain
| | - Sara Vigil‐Vázquez
- Sevicio de NeonatologíaHospital General Universitario Gregorio MarañónMadridSpain
| | - Mónica Vázquez
- Unidad de Biología ViralCentro Nacional de Microbiología, Instituto de Salud Carlos IIIMadridSpain
| | - Angela Manzanares
- Servicio de PediatríaHospital General Universitario Gregorio MarañónMadridSpain
| | - Olga Cano
- Unidad de Biología ViralCentro Nacional de Microbiología, Instituto de Salud Carlos IIIMadridSpain
| | - Roberto Alonso
- Departamento de Microbiología Clínica y Enfermedades InfecciosasHospital General Universitario Gregorio MarañónMadridSpain
| | - Daniel Sepúlveda‐Crespo
- Unidad de Infección Viral e InmunidadCentro Nacional de Microbiología, Instituto de Salud Carlos III, MajadahondaMadridSpain
| | - Laura Tarancón‐Díez
- Laboratorio de InmunoBiología Molecular, Sección de InmunologíaHospital General Universitario Gregorio MarañónMadridSpain
| | - María Ángeles Muñoz‐Fernández
- Laboratorio de InmunoBiología Molecular, Sección de InmunologíaHospital General Universitario Gregorio MarañónMadridSpain
- HIV‐HGM BioBankMadridSpain
| | - Mar Muñoz‐Chapuli
- Departamento de Obstetricia y GinecologíaHospital General Universitario Gregorio MarañónMadridSpain
| | - Salvador Resino
- Unidad de Infección Viral e InmunidadCentro Nacional de Microbiología, Instituto de Salud Carlos III, MajadahondaMadridSpain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC)Instituto de Salud Carlos IIIMadridSpain
| | - Maria Luisa Navarro
- Grupo de Investigación en Infectología PediátricaInstituto de Investigación Sanitaria Gregorio MarañónMadridSpain
- Servicio de PediatríaHospital General Universitario Gregorio MarañónMadridSpain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC)Instituto de Salud Carlos IIIMadridSpain
| | - Isidoro Martinez
- Unidad de Infección Viral e InmunidadCentro Nacional de Microbiología, Instituto de Salud Carlos III, MajadahondaMadridSpain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC)Instituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
45
|
Nziza N, Jung W, Mendu M, Chen T, McNamara RP, Fortune SM, Franken KLMC, Ottenhoff THM, Bryson B, Ngonzi J, Bebell LM, Alter G. Maternal HIV infection drives altered placental Mtb-specific antibody transfer. Front Microbiol 2023; 14:1171990. [PMID: 37228375 PMCID: PMC10203169 DOI: 10.3389/fmicb.2023.1171990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/17/2023] [Indexed: 05/27/2023] Open
Abstract
Introduction Placental transfer of maternal antibodies is essential for neonatal immunity over the first months of life. In the setting of maternal HIV infection, HIV-exposed uninfected (HEU) infants are at higher risk of developing severe infections, including active tuberculosis (TB). Given our emerging appreciation for the potential role of antibodies in the control of Mycobacterium tuberculosis (Mtb), the bacteria that causes TB, here we aimed to determine whether maternal HIV status altered the quality of Mtb-specific placental antibody transfer. Methods Antigen-specific antibody systems serology was performed to comprehensively characterize the Mtb-specific humoral immune response in maternal and umbilical cord blood from HIV infected and uninfected pregnant people in Uganda. Results Significant differences were noted in overall antibody profiles in HIV positive and negative maternal plasma, resulting in heterogeneous transfer of Mtb-specific antibodies. Altered antibody transfer in HIV infected dyads was associated with impaired binding to IgG Fc-receptors, which was directly linked to HIV viral loads and CD4 counts. Conclusions These results highlight the importance of maternal HIV status on antibody transfer, providing clues related to alterations in transferred maternal immunity that may render HEU infants more vulnerable to TB than their HIV-unexposed peers.
Collapse
Affiliation(s)
- Nadege Nziza
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
| | - Wonyeong Jung
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
| | - Maanasa Mendu
- Department of Molecular and Cellular Biology, Harvard University, Boston, MA, United States
| | - Tina Chen
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
| | - Ryan P. McNamara
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
| | - Sarah M. Fortune
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Kees L. M. C. Franken
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Bryan Bryson
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Joseph Ngonzi
- Department of Obstetrics and Gynecology, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Lisa M. Bebell
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, United States
- Medical Practice Evaluation Center, Massachusetts General Hospital, Boston, MA, United States
- Center for Global Health, Massachusetts General Hospital, Boston, MA, United States
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
46
|
Adhikari EH, Lu P, Kang YJ, McDonald AR, Pruszynski JE, Bates TA, McBride SK, Trank-Greene M, Tafesse FG, Lu LL. Diverging maternal and infant cord antibody functions from SARS-CoV-2 infection and vaccination in pregnancy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.01.538955. [PMID: 37205338 PMCID: PMC10187183 DOI: 10.1101/2023.05.01.538955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Immunization in pregnancy is a critical tool that can be leveraged to protect the infant with an immature immune system but how vaccine-induced antibodies transfer to the placenta and protect the maternal-fetal dyad remains unclear. Here, we compare matched maternal-infant cord blood from individuals who in pregnancy received mRNA COVID-19 vaccine, were infected by SARS-CoV-2, or had the combination of these two immune exposures. We find that some but not all antibody neutralizing activities and Fc effector functions are enriched with vaccination compared to infection. Preferential transport to the fetus of Fc functions and not neutralization is observed. Immunization compared to infection enriches IgG1-mediated antibody functions with changes in antibody post-translational sialylation and fucosylation that impact fetal more than maternal antibody functional potency. Thus, vaccine enhanced antibody functional magnitude, potency and breadth in the fetus are driven more by antibody glycosylation and Fc effector functions compared to maternal responses, highlighting prenatal opportunities to safeguard newborns as SARS-CoV-2 becomes endemic.
Collapse
Affiliation(s)
- Emily H. Adhikari
- Division of Maternal-Fetal Medicine and Department of Obstetrics and Gynecology, UTSW Medical Center, Dallas, TX
- Parkland Health, Dallas TX
| | - Pei Lu
- Division of Infectious Diseases and Geographic Medicine and Department of Internal Medicine, UTSW Medical Center, Dallas, TX
| | - Ye jin Kang
- Division of Infectious Diseases and Geographic Medicine and Department of Internal Medicine, UTSW Medical Center, Dallas, TX
| | - Ann R. McDonald
- Division of Infectious Diseases and Geographic Medicine and Department of Internal Medicine, UTSW Medical Center, Dallas, TX
| | - Jessica E. Pruszynski
- Division of Maternal-Fetal Medicine and Department of Obstetrics and Gynecology, UTSW Medical Center, Dallas, TX
| | - Timothy A. Bates
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| | - Savannah K. McBride
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| | - Mila Trank-Greene
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| | - Fikadu G. Tafesse
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| | - Lenette L. Lu
- Parkland Health, Dallas TX
- Division of Infectious Diseases and Geographic Medicine and Department of Internal Medicine, UTSW Medical Center, Dallas, TX
- Department of Immunology, UTSW Medical Center, Dallas, TX
| |
Collapse
|
47
|
Zelini P, d'Angelo P, Zavaglio F, Soleymaninejadian E, Mariani L, Perotti F, Dominoni M, Tonello S, Sainaghi P, Minisini R, Apostolo D, Lilleri D, Spinillo A, Baldanti F. Inflammatory and Immune Responses during SARS-CoV-2 Infection in Vaccinated and Non-Vaccinated Pregnant Women and Their Newborns. Pathogens 2023; 12:pathogens12050664. [PMID: 37242334 DOI: 10.3390/pathogens12050664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Pregnant women are more susceptible to severe disease associated with SARS-CoV-2 infection. We performed a prospective study to analyze the inflammatory and immune profile after SARS-CoV-2 infection occurring in vaccinated or non-vaccinated pregnant women and their newborns. METHODS Twenty-five pregnant women with SARS-CoV-2 infection were enrolled, and sixteen cord blood samples were obtained at delivery. RESULTS We observed that IL-1β, TNF-α, Eotaxin, MIB-1β, VEGF, IL-15, IL-2, IL-5, IL-9, IL-10 and IL-1ra levels were significantly higher in vaccinated than non-vaccinated mothers. Furthermore, the newborns of the vaccinated mothers produced higher levels of IL-7, IL-5 and IL-12 compared to the newborns of non-vaccinated mothers. Anti-Spike (S) IgG levels were significantly higher in all vaccinated mothers and their newborns compared to the non-vaccinated group. We found that 87.5% of vaccinated women and 66.6% of non-vaccinated women mounted an S-specific T-cell response quantified by ELISpot assay. Moreover, 75.0% of vaccinated mothers and 38.4% of non-vaccinated mothers showed S-specific CD4+ T-cell proliferative response. The T-helper subset response was restricted to CD4+ Th1 in both vaccinated and non-vaccinated women. CONCLUSION A higher level of cytokines, IgG antibodies and memory T cells was noted in the vaccinated women. Furthermore, the maternal IgG antibody trans-placental transfer occurred more frequently in vaccinated mothers and may protect the newborn.
Collapse
Affiliation(s)
- Paola Zelini
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Piera d'Angelo
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Obstetrics and Gynecology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Federica Zavaglio
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Ehsan Soleymaninejadian
- Department of Clinical, Surgical, Diagnostics and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| | - Liliana Mariani
- Obstetrics and Gynecology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Francesca Perotti
- Obstetrics and Gynecology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Mattia Dominoni
- Obstetrics and Gynecology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Stelvio Tonello
- Immunoreumatology Laboratory, Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Piemonte Orientale, 28100 Novara, Italy
- Internal Medicine Laboratory, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Pierpaolo Sainaghi
- Immunoreumatology Laboratory, Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Piemonte Orientale, 28100 Novara, Italy
- Internal Medicine Laboratory, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Rosalba Minisini
- Internal Medicine Laboratory, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Daria Apostolo
- Immunoreumatology Laboratory, Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Piemonte Orientale, 28100 Novara, Italy
- Internal Medicine Laboratory, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Daniele Lilleri
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Arsenio Spinillo
- Obstetrics and Gynecology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Department of Clinical, Surgical, Diagnostics and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| | - Fausto Baldanti
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Department of Clinical, Surgical, Diagnostics and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
48
|
Habel JR, Chua BY, Kedzierski L, Selva KJ, Damelang T, Haycroft ER, Nguyen TH, Koay HF, Nicholson S, McQuilten HA, Jia X, Allen LF, Hensen L, Zhang W, van de Sandt CE, Neil JA, Pragastis K, Lau JS, Jumarang J, Allen EK, Amanant F, Krammer F, Wragg KM, Juno JA, Wheatley AK, Tan HX, Pell G, Walker S, Audsley J, Reynaldi A, Thevarajan I, Denholm JT, Subbarao K, Davenport MP, Hogarth PM, Godfrey DI, Cheng AC, Tong SY, Bond K, Williamson DA, McMahon JH, Thomas PG, Pannaraj PS, James F, Holmes NE, Smibert OC, Trubiano JA, Gordon CL, Chung AW, Whitehead CL, Kent SJ, Lappas M, Rowntree LC, Kedzierska K. Immune profiling of SARS-CoV-2 infection during pregnancy reveals NK cell and γδ T cell perturbations. JCI Insight 2023; 8:e167157. [PMID: 37036008 PMCID: PMC10132165 DOI: 10.1172/jci.insight.167157] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/15/2023] [Indexed: 04/11/2023] Open
Abstract
Pregnancy poses a greater risk for severe COVID-19; however, underlying immunological changes associated with SARS-CoV-2 during pregnancy are poorly understood. We defined immune responses to SARS-CoV-2 in unvaccinated pregnant and nonpregnant women with acute and convalescent COVID-19, quantifying 217 immunological parameters. Humoral responses to SARS-CoV-2 were similar in pregnant and nonpregnant women, although our systems serology approach revealed distinct antibody and FcγR profiles between pregnant and nonpregnant women. Cellular analyses demonstrated marked differences in NK cell and unconventional T cell activation dynamics in pregnant women. Healthy pregnant women displayed preactivated NK cells and γδ T cells when compared with healthy nonpregnant women, which remained unchanged during acute and convalescent COVID-19. Conversely, nonpregnant women had prototypical activation of NK and γδ T cells. Activation of CD4+ and CD8+ T cells and T follicular helper cells was similar in SARS-CoV-2-infected pregnant and nonpregnant women, while antibody-secreting B cells were increased in pregnant women during acute COVID-19. Elevated levels of IL-8, IL-10, and IL-18 were found in pregnant women in their healthy state, and these cytokine levels remained elevated during acute and convalescent COVID-19. Collectively, we demonstrate perturbations in NK cell and γδ T cell activation in unvaccinated pregnant women with COVID-19, which may impact disease progression and severity during pregnancy.
Collapse
Affiliation(s)
- Jennifer R. Habel
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Brendon Y. Chua
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
| | - Lukasz Kedzierski
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Kevin J. Selva
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Timon Damelang
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Ebene R. Haycroft
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Thi H.O. Nguyen
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Hui-Fern Koay
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Suellen Nicholson
- Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Hayley A. McQuilten
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Xiaoxiao Jia
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Lilith F. Allen
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Luca Hensen
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Wuji Zhang
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Carolien E. van de Sandt
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jessica A. Neil
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Katherine Pragastis
- Department of Infectious Diseases, Alfred Health, Monash University, Melbourne, Victoria, Australia
| | - Jillian S.Y. Lau
- Department of Infectious Diseases, Alfred Health, Monash University, Melbourne, Victoria, Australia
- Department of Infectious Diseases, Eastern Health, Box Hill, Victoria, Australia
| | - Jaycee Jumarang
- Division of Infectious Diseases, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - E. Kaitlynn Allen
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Fatima Amanant
- Department of Microbiology, and
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Kathleen M. Wragg
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jennifer A. Juno
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Adam K. Wheatley
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, Victoria, Australia
| | - Hyon-Xhi Tan
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Gabrielle Pell
- Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Susan Walker
- Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Jennifer Audsley
- Department of Infectious Diseases, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Irani Thevarajan
- Department of Infectious Diseases, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Justin T. Denholm
- Department of Infectious Diseases, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Miles P. Davenport
- Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - P. Mark Hogarth
- Immune Therapies Laboratory, Burnet Institute, Melbourne, Victoria, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Clinical Pathology, University of Melbourne, Parkville, Victoria, Australia
| | - Dale I. Godfrey
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Allen C. Cheng
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Infection Prevention and Healthcare Epidemiology Unit, Alfred Health, and Monash Infectious Diseases, Monash Health, Melbourne, Victoria, Australia
| | - Steven Y.C. Tong
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Katherine Bond
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Microbiology, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Deborah A. Williamson
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Microbiology, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - James H. McMahon
- Department of Infectious Diseases, Alfred Health, Monash University, Melbourne, Victoria, Australia
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Pia S. Pannaraj
- Division of Infectious Diseases, Children’s Hospital Los Angeles, Los Angeles, California, USA
- Departments of Pediatrics, Molecular Microbiology and Immunology, Keck School of Medicine, UCLA, Los Angeles, California, USA
| | - Fiona James
- Department of Infectious Diseases, Austin Health, Heidelberg, Victoria, Australia
| | - Natasha E. Holmes
- Department of Infectious Diseases, Austin Health, Heidelberg, Victoria, Australia
- Department of Critical Care, University of Melbourne, Parkville, Victoria, Australia
- Data Analytics Research and Evaluation Centre, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
- Centre for Antibiotic Allergy and Research, Department of Infectious Diseases, Austin Health, Heidelberg, Victoria, Australia
| | - Olivia C. Smibert
- Departments of Pediatrics, Molecular Microbiology and Immunology, Keck School of Medicine, UCLA, Los Angeles, California, USA
- Department of Infectious Diseases, Austin Health, Heidelberg, Victoria, Australia
- Centre for Antibiotic Allergy and Research, Department of Infectious Diseases, Austin Health, Heidelberg, Victoria, Australia
- Department of Infectious Diseases, and
- National Centre for Infections in Cancer, Peter McCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Jason A. Trubiano
- Centre for Antibiotic Allergy and Research, Department of Infectious Diseases, Austin Health, Heidelberg, Victoria, Australia
- Department of Infectious Diseases, and
- National Centre for Infections in Cancer, Peter McCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Medicine (Austin Health), University of Melbourne, Heidelberg, Victoria, Australia
| | - Claire L. Gordon
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Infectious Diseases, Austin Health, Heidelberg, Victoria, Australia
| | - Amy W. Chung
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Clare L. Whitehead
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
- Pregnancy Research Centre, Royal Women’s Hospital, Parkville, Victoria, Australia
| | - Stephen J. Kent
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, Victoria, Australia
- Melbourne Sexual Health Centre, Infectious Diseases Department, Alfred Health, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Martha Lappas
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - Louise C. Rowntree
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
| |
Collapse
|
49
|
Back JH, Kim SY, Gu MB, Kim HJ, Lee KN, Lee JE, Park KH. Proteomic analysis of plasma to identify novel biomarkers for intra-amniotic infection and/or inflammation in preterm premature rupture of membranes. Sci Rep 2023; 13:5658. [PMID: 37024561 PMCID: PMC10079851 DOI: 10.1038/s41598-023-32884-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 04/04/2023] [Indexed: 04/08/2023] Open
Abstract
To identify potential plasma biomarkers associated with microbial invasion of the amniotic cavity (MIAC) and/or intraamniotic inflammation (IAI) in women with preterm premature rupture of membranes (PPROM). This retrospective cohort study included 182 singleton pregnant women with PPROM (23-33 weeks) who underwent amniocentesis. Plasma samples; all subjects were chosen from these participants and were analyzed using label-free liquid chromatography-tandem mass spectrometry for proteome profiling using a nested case-control study design (cases with MIAC/IAI vs. non-MIAC/IAI controls [n = 9 each]). Three identified target molecules for MIAC/IAI were further verified by ELISA in the study cohort (n = 182). Shotgun proteomic analysis revealed 17 differentially expressed proteins (P < 0.05) in the plasma of MIAC/IAI cases. In particular, the levels of FCGR3A and haptoglobin, but not LRP1, were found to be increased in the plasma of patients with MIAC, IAI, and both MIAC/IAI compared with those without these conditions. Moreover, these differences remained significant after adjusting for gestational age at sampling. The area under the curves of plasma FCGR3A and haptoglobin ranged within 0.59-0.65 with respect to each of the three outcome measures. Plasma FCGR3A and haptoglobin were identified as potential independent biomarkers for less-invasively detecting MIAC/IAI in women with PPROM.
Collapse
Affiliation(s)
- Ji Hyun Back
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Korea
- Biomedical Research Division, Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Korea
| | - So Yeon Kim
- Department of Obstetrics and Gynecology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Man Bock Gu
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Korea
| | - Hyeon Ji Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 82, Gumi-Ro 173 Beon-Gil, Bundang-Gu, Seongnam, 463-707, Korea
| | - Kyong-No Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 82, Gumi-Ro 173 Beon-Gil, Bundang-Gu, Seongnam, 463-707, Korea
| | - Ji Eun Lee
- Biomedical Research Division, Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Korea.
| | - Kyo Hoon Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 82, Gumi-Ro 173 Beon-Gil, Bundang-Gu, Seongnam, 463-707, Korea.
| |
Collapse
|
50
|
Gomez-Lopez N, Romero R, Escobar MF, Carvajal JA, Echavarria MP, Albornoz LL, Nasner D, Miller D, Gallo DM, Galaz J, Arenas-Hernandez M, Bhatti G, Done B, Zambrano MA, Ramos I, Fernandez PA, Posada L, Chaiworapongsa T, Jung E, Garcia-Flores V, Suksai M, Gotsch F, Bosco M, Than NG, Tarca AL. Pregnancy-specific responses to COVID-19 revealed by high-throughput proteomics of human plasma. COMMUNICATIONS MEDICINE 2023; 3:48. [PMID: 37016066 PMCID: PMC10071476 DOI: 10.1038/s43856-023-00268-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 03/03/2023] [Indexed: 04/06/2023] Open
Abstract
BACKGROUND Pregnant women are at greater risk of adverse outcomes, including mortality, as well as obstetrical complications resulting from COVID-19. However, pregnancy-specific changes that underlie such worsened outcomes remain unclear. METHODS Plasma samples were collected from pregnant women and non-pregnant individuals (male and female) with (n = 72 pregnant, 52 non-pregnant) and without (n = 29 pregnant, 41 non-pregnant) COVID-19. COVID-19 patients were grouped as asymptomatic, mild, moderate, severe, or critically ill according to NIH classifications. Proteomic profiling of 7,288 analytes corresponding to 6,596 unique protein targets was performed using the SOMAmer platform. RESULTS Herein, we profile the plasma proteome of pregnant and non-pregnant COVID-19 patients and controls and show alterations that display a dose-response relationship with disease severity; yet, such proteomic perturbations are dampened during pregnancy. In both pregnant and non-pregnant state, the proteome response induced by COVID-19 shows enrichment of mediators implicated in cytokine storm, endothelial dysfunction, and angiogenesis. Shared and pregnancy-specific proteomic changes are identified: pregnant women display a tailored response that may protect the conceptus from heightened inflammation, while non-pregnant individuals display a stronger response to repel infection. Furthermore, the plasma proteome can accurately identify COVID-19 patients, even when asymptomatic or with mild symptoms. CONCLUSION This study represents the most comprehensive characterization of the plasma proteome of pregnant and non-pregnant COVID-19 patients. Our findings emphasize the distinct immune modulation between the non-pregnant and pregnant states, providing insight into the pathogenesis of COVID-19 as well as a potential explanation for the more severe outcomes observed in pregnant women.
Collapse
Affiliation(s)
- Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA.
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA.
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA.
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA.
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA.
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA.
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA.
- Detroit Medical Center, Detroit, MI, USA.
| | - María Fernanda Escobar
- Departamento de Ginecología y Obstetricia, Fundación Valle del Lili, Cali, Colombia
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Javier Andres Carvajal
- Departamento de Ginecología y Obstetricia, Fundación Valle del Lili, Cali, Colombia
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Maria Paula Echavarria
- Departamento de Ginecología y Obstetricia, Fundación Valle del Lili, Cali, Colombia
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Ludwig L Albornoz
- Departamento de Laboratorio Clínico y Patología, Fundación Valle del Lili, Cali, Colombia
- Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Daniela Nasner
- Centro de Investigaciones Clínicas, Fundación Valle del Lili, Cali, Colombia
| | - Derek Miller
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Dahiana M Gallo
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jose Galaz
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcia Arenas-Hernandez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Gaurav Bhatti
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Bogdan Done
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Maria Andrea Zambrano
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Isabella Ramos
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Paula Andrea Fernandez
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Leandro Posada
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Tinnakorn Chaiworapongsa
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Eunjung Jung
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Valeria Garcia-Flores
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Manaphat Suksai
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Francesca Gotsch
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Mariachiara Bosco
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nandor Gabor Than
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
- Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary
- Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
- Genesis Theranostix Group, Budapest, Hungary
| | - Adi L Tarca
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA.
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA.
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, USA.
| |
Collapse
|