1
|
Mane V, Mehta R, Alvarez N, Sharma V, Park S, Fox A, DeCarlo C, Yang X, Perlin DS, Powell RLR. In vivo antiviral efficacy of LCTG-002, a pooled, purified human milk secretory IgA product, against SARS-CoV-2 in a murine model of COVID-19. Hum Vaccin Immunother 2024; 20:2303226. [PMID: 38251677 PMCID: PMC10807469 DOI: 10.1080/21645515.2024.2303226] [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: 09/25/2023] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
Immunoglobulin A (IgA) is the most abundant antibody (Ab) in human mucosae, with secretory form (sIgA) being dominant and uniquely stable. sIgA is challenging to produce recombinantly but is naturally found in human milk, which could be considered a global resource for this biologic, justifying its development as a mucosal therapeutic. Presently, SARS-CoV-2 was utilized as a model mucosal pathogen, and methods were developed to efficiently extract human milk sIgA from donors who were naïve to SARS-CoV-2 or had recovered from infection that elicited high-titer anti-SARS-CoV-2 Spike sIgA in their milk (pooled to make LCTG-002). Mass spectrometry determined that proteins with a relative abundance of 1% or greater were all associated with sIgA. Western blot demonstrated that all batches consisted predominantly of sIgA. Compared to control IgA, LCTG-002 demonstrated significantly higher Spike binding (mean endpoint of 0.87 versus 5.87). LCTG-002 was capable of blocking the Spike receptor-binding domain - angiotensin-converting enzyme 2 (ACE2) interaction with significantly greater potency compared to control (mean LCTG-002 IC50 154ug/mL versus 50% inhibition not achieved for control), and exhibited significant neutralization activity against Spike-pseudotyped virus infection (mean LCTG-002 IC50 49.8ug/mL versus 114.5ug/mL for control). LCTG-002 was tested for its capacity to reduce viral lung burden in K18+hACE2 transgenic mice inoculated with SARS-CoV-2. LCTG-002 significantly reduced SARS-CoV-2 titers compared to control when administered at 0.25 mg/day or 1 mg/day, with a maximum TCID50 reduction of 4.9 logs. This innovative study demonstrates that LCTG-002 is highly pure and efficacious in vivo, supporting further development of milk-derived, polyclonal sIgA therapeutics.
Collapse
Affiliation(s)
- Viraj Mane
- Lactiga US, Inc. 675 US-1, North Brunswick, NJ, USA
| | - Rikin Mehta
- Lactiga US, Inc. 675 US-1, North Brunswick, NJ, USA
| | - Nadine Alvarez
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Vijeta Sharma
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Steven Park
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Alisa Fox
- Icahn School of Medicine at Mount Sinai, Department of Medicine, Division of Infectious Diseases, New York, NY, USA
| | - Claire DeCarlo
- Icahn School of Medicine at Mount Sinai, Department of Medicine, Division of Infectious Diseases, New York, NY, USA
| | - Xiaoqi Yang
- Icahn School of Medicine at Mount Sinai, Department of Medicine, Division of Infectious Diseases, New York, NY, USA
| | - David S. Perlin
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Rebecca L. R. Powell
- Icahn School of Medicine at Mount Sinai, Department of Medicine, Division of Infectious Diseases, New York, NY, USA
| |
Collapse
|
2
|
Hochmayr C, Winkler I, Hammerl M, Höller A, Huber E, Urbanek M, Kiechl-Kohlendorfer U, Griesmaier E, Posod A. Factors Influencing Breast Milk Antibody Titers during the Coronavirus Disease 2019 Pandemic: An Observational Study. Nutrients 2024; 16:2320. [PMID: 39064762 PMCID: PMC11280407 DOI: 10.3390/nu16142320] [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: 06/25/2024] [Revised: 07/09/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
The COVID-19 pandemic has highlighted the role of breastfeeding in providing passive immunity to infants via specific anti-SARS-CoV-2 antibodies in breast milk. We aimed to quantify these antibodies across different lactation stages and identify influencing factors. This prospective study involved mother-child dyads from Innsbruck University Hospital, Austria, with a positive maternal SARS-CoV-2 test during pregnancy or peripartum between 2020 and 2023. We collected breast milk samples at various lactation stages and analyzed anti-Spike S1 receptor-binding domain (S1RBD) immunoglobulins (Ig). Maternal and neonatal data were obtained from interviews and medical records. This study included 140 mothers and 144 neonates. Anti-S1RBD-IgA (72.0%), -IgG (86.0%), and -IgM (41.7%) were highly present in colostrum and decreased as milk matured. Mothers with natural infection and vaccination exhibited higher anti-S1RBD-IgA and -IgG titers in all milk stages. Mothers with moderate to severe infections had higher concentrations of anti-S1RBD-IgA and -IgG in transitional milk and higher anti-S1RBD-IgA and -IgM in mature milk compared to those with mild or asymptomatic infections. Variations in antibody responses were also observed with preterm birth and across different virus waves. This study demonstrates the dynamic nature of breast milk Ig and underscores the importance of breastfeeding during a pandemic.
Collapse
Affiliation(s)
- Christoph Hochmayr
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Ira Winkler
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Marlene Hammerl
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Alexander Höller
- Division for Nutrition and Dietetics, University Hospital Innsbruck, 6020 Innsbruck, Austria
- Institute of Public Health, Medical Decision Making and Health Technology Assessment, Department of Public Health, Health Services Research and Health Technology Assessment, UMIT TIROL—University for Health Sciences and Technology, 6060 Hall in Tirol, Austria
| | - Eva Huber
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Martina Urbanek
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Ursula Kiechl-Kohlendorfer
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Elke Griesmaier
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Anna Posod
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| |
Collapse
|
3
|
Mattar SB, Celedon PAF, Leony LM, Vasconcelos LDCM, Sampaio DD, Marchini FK, Morello LG, Lin VH, Crestani S, Camelier AA, Meireles AC, de Oliveira Junior ALF, Bandeira AC, Macedo YSF, Duarte AO, Pavan TBS, de Siqueira IC, Santos FLN. Comprehensive Study of the IBMP ELISA IgA/IgM/IgG COVID-19 Kit for SARS-CoV-2 Antibody Detection. Diagnostics (Basel) 2024; 14:1514. [PMID: 39061652 PMCID: PMC11276192 DOI: 10.3390/diagnostics14141514] [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: 06/16/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
COVID-19 laboratory diagnosis primarily relies on molecular tests, highly sensitive during early infection stages with high viral loads. As the disease progresses, sensitivity decreases, requiring antibody detection. Since the beginning of the pandemic, serological tests have been developed and made available in Brazil, but their diagnostic performance varies. This study evaluated the IBMP ELISA IgA/IgM/IgG COVID-19 kit performance in detecting SARS-CoV-2 antibodies. A total of 90 samples, including 64 from COVID-19 patients and 26 pre-pandemic donors, were assessed based on time post symptom onset (0-7, 8-14, and 15-21 days). The kit showed 61% sensitivity, 100% specificity, and 72% accuracy overall. Sensitivity varied with time, being 25%, 57%, and 96% for 0-7, 8-14, and 15-21 days, respectively. Similar variations were noted in other commercial tests. The Gold ELISA COVID-19 (IgG/IgM) had sensitivities of 31%, 71%, and 100%, while the Anti-SARS-CoV-2 NCP ELISA (IgG) and Anti-SARS-CoV-2 NCP ELISA (IgM) showed varying sensitivities. The IBMP ELISA kit displayed high diagnostic capability, especially as the disease progressed, complementing COVID-19 diagnosis. Reproducibility assessment revealed minimal systematic and analytical errors. In conclusion, the IBMP ELISA IgA/IgM/IgG COVID-19 kit is a robust tool for detecting anti-SARS-CoV-2 antibodies, increasing in efficacy over the disease course, and minimizing false negatives in RT-PCR COVID-19 diagnosis.
Collapse
Affiliation(s)
- Sibelle Botogosque Mattar
- Molecular Biology Institute of Paraná (IBMP), Curitiba 81350-010, PR, Brazil; (S.B.M.); (P.A.F.C.); (F.K.M.); (L.G.M.); (V.H.L.); (S.C.)
| | - Paola Alejandra Fiorani Celedon
- Molecular Biology Institute of Paraná (IBMP), Curitiba 81350-010, PR, Brazil; (S.B.M.); (P.A.F.C.); (F.K.M.); (L.G.M.); (V.H.L.); (S.C.)
- Interdisciplinary Research Group in Biotechnology and Epidemiology of Infectious Diseases (GRUPIBE), Gonçalo Moniz Institute, Oswaldo Cruz Foundation-Bahia (FIOCRUZ-BA), Salvador 402596-710, BA, Brazil; (L.M.L.); (L.d.C.M.V.); (D.D.S.); (T.B.S.P.); (I.C.d.S.)
| | - Leonardo Maia Leony
- Interdisciplinary Research Group in Biotechnology and Epidemiology of Infectious Diseases (GRUPIBE), Gonçalo Moniz Institute, Oswaldo Cruz Foundation-Bahia (FIOCRUZ-BA), Salvador 402596-710, BA, Brazil; (L.M.L.); (L.d.C.M.V.); (D.D.S.); (T.B.S.P.); (I.C.d.S.)
- Advanced Public Health Laboratory, Gonçalo Moniz Institute, Oswaldo Cruz Foundation-Bahia (FIOCRUZ-BA), Salvador 402596-710, BA, Brazil
| | - Larissa de Carvalho Medrado Vasconcelos
- Interdisciplinary Research Group in Biotechnology and Epidemiology of Infectious Diseases (GRUPIBE), Gonçalo Moniz Institute, Oswaldo Cruz Foundation-Bahia (FIOCRUZ-BA), Salvador 402596-710, BA, Brazil; (L.M.L.); (L.d.C.M.V.); (D.D.S.); (T.B.S.P.); (I.C.d.S.)
- Advanced Public Health Laboratory, Gonçalo Moniz Institute, Oswaldo Cruz Foundation-Bahia (FIOCRUZ-BA), Salvador 402596-710, BA, Brazil
| | - Daniel Dias Sampaio
- Interdisciplinary Research Group in Biotechnology and Epidemiology of Infectious Diseases (GRUPIBE), Gonçalo Moniz Institute, Oswaldo Cruz Foundation-Bahia (FIOCRUZ-BA), Salvador 402596-710, BA, Brazil; (L.M.L.); (L.d.C.M.V.); (D.D.S.); (T.B.S.P.); (I.C.d.S.)
| | - Fabricio Klerynton Marchini
- Molecular Biology Institute of Paraná (IBMP), Curitiba 81350-010, PR, Brazil; (S.B.M.); (P.A.F.C.); (F.K.M.); (L.G.M.); (V.H.L.); (S.C.)
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation-Paraná (FIOCRUZ-PR), Curitiba 81350-010, PR, Brazil
| | - Luis Gustavo Morello
- Molecular Biology Institute of Paraná (IBMP), Curitiba 81350-010, PR, Brazil; (S.B.M.); (P.A.F.C.); (F.K.M.); (L.G.M.); (V.H.L.); (S.C.)
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation-Paraná (FIOCRUZ-PR), Curitiba 81350-010, PR, Brazil
| | - Vanessa Hoysan Lin
- Molecular Biology Institute of Paraná (IBMP), Curitiba 81350-010, PR, Brazil; (S.B.M.); (P.A.F.C.); (F.K.M.); (L.G.M.); (V.H.L.); (S.C.)
| | - Sandra Crestani
- Molecular Biology Institute of Paraná (IBMP), Curitiba 81350-010, PR, Brazil; (S.B.M.); (P.A.F.C.); (F.K.M.); (L.G.M.); (V.H.L.); (S.C.)
| | | | - André Costa Meireles
- Aliança D’Or Hospital, Salvador 41920-180, BA, Brazil; (A.A.C.); (A.C.M.); (A.L.F.d.O.J.)
| | | | | | - Yasmin Santos Freitas Macedo
- Laboratory of Investigation in Global Health and Neglected Diseases, Gonçalo Moniz Institute, Oswaldo Cruz Foundation-Bahia (FIOCRUZ-BA), Salvador 402596-710, BA, Brazil; (Y.S.F.M.); (A.O.D.)
| | - Alan Oliveira Duarte
- Laboratory of Investigation in Global Health and Neglected Diseases, Gonçalo Moniz Institute, Oswaldo Cruz Foundation-Bahia (FIOCRUZ-BA), Salvador 402596-710, BA, Brazil; (Y.S.F.M.); (A.O.D.)
| | - Tycha Bianca Sabaini Pavan
- Interdisciplinary Research Group in Biotechnology and Epidemiology of Infectious Diseases (GRUPIBE), Gonçalo Moniz Institute, Oswaldo Cruz Foundation-Bahia (FIOCRUZ-BA), Salvador 402596-710, BA, Brazil; (L.M.L.); (L.d.C.M.V.); (D.D.S.); (T.B.S.P.); (I.C.d.S.)
- Advanced Public Health Laboratory, Gonçalo Moniz Institute, Oswaldo Cruz Foundation-Bahia (FIOCRUZ-BA), Salvador 402596-710, BA, Brazil
| | - Isadora Cristina de Siqueira
- Interdisciplinary Research Group in Biotechnology and Epidemiology of Infectious Diseases (GRUPIBE), Gonçalo Moniz Institute, Oswaldo Cruz Foundation-Bahia (FIOCRUZ-BA), Salvador 402596-710, BA, Brazil; (L.M.L.); (L.d.C.M.V.); (D.D.S.); (T.B.S.P.); (I.C.d.S.)
- Laboratory of Investigation in Global Health and Neglected Diseases, Gonçalo Moniz Institute, Oswaldo Cruz Foundation-Bahia (FIOCRUZ-BA), Salvador 402596-710, BA, Brazil; (Y.S.F.M.); (A.O.D.)
- Integrated Translational Program in Chagas Disease from FIOCRUZ (Fio-Chagas), Oswaldo Cruz Foundation-Rio de Janeiro (FIOCRUZ-RJ), Rio de Janeiro 21040-360, RJ, Brazil
| | - Fred Luciano Neves Santos
- Interdisciplinary Research Group in Biotechnology and Epidemiology of Infectious Diseases (GRUPIBE), Gonçalo Moniz Institute, Oswaldo Cruz Foundation-Bahia (FIOCRUZ-BA), Salvador 402596-710, BA, Brazil; (L.M.L.); (L.d.C.M.V.); (D.D.S.); (T.B.S.P.); (I.C.d.S.)
- Advanced Public Health Laboratory, Gonçalo Moniz Institute, Oswaldo Cruz Foundation-Bahia (FIOCRUZ-BA), Salvador 402596-710, BA, Brazil
- Integrated Translational Program in Chagas Disease from FIOCRUZ (Fio-Chagas), Oswaldo Cruz Foundation-Rio de Janeiro (FIOCRUZ-RJ), Rio de Janeiro 21040-360, RJ, Brazil
| |
Collapse
|
4
|
Muñoz-Gómez MJ, Martin-Vicente M, Vigil-Vazquez S, Carrasco I, Lobo AH, Mas V, Vázquez M, Manzanares A, Cano O, Zamora C, Alonso R, Sepulveda-Crespo D, Tarancon-Diez L, Muñoz-Fernández MÁ, Muñoz-Chapuli M, Resino S, Navarro ML, Martinez I. IgG antibody levels against the SARS-CoV-2 spike protein in mother-child dyads after COVID-19 vaccination. Infection 2024; 52:813-824. [PMID: 37898587 DOI: 10.1007/s15010-023-02111-x] [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: 04/27/2023] [Accepted: 10/08/2023] [Indexed: 10/30/2023]
Abstract
PURPOSE We aimed to assess IgG antibodies against the SARS-CoV-2 spike protein (anti-SARS-CoV-2 S IgG) in vaccinated mothers and their infants at delivery and 2-3 months of age. METHODS We conducted a prospective study on mothers who received at least one dose of the COVID-19 vaccine (Pfizer-BNT162b2, Moderna mRNA-1273, or Oxford-AstraZeneca ChAdOx1-S) during pregnancy and on their infants. The baseline was at the time of delivery (n = 93), and the end of follow-up was 2 to 3 months post-partum (n = 53). Serum anti-SARS-CoV-2 S IgG titers and ACE2 binding inhibition levels were quantified by immunoassays. RESULTS Mothers and infants had high anti-SARS-CoV-2 S IgG titers against the B.1 lineage at birth. However, while antibody titers were maintained at 2-3 months post-partum in mothers, they decreased significantly in infants (p < 0.001). Positive and significant correlations were found between anti-SARS-CoV-2 S IgG titers and ACE2-binding inhibition levels in mothers and infants at birth and 2-3 months post-partum (r > 0.8, p < 0.001). Anti-S antibodies were also quantified for the Omicron variant at 2-3 months post-partum. The antibody titers against Omicron were significantly lower in mothers and infants than those against B.1 (p < 0.001). Again, a positive correlation was observed for Omicron between IgG titers and ACE2-binding inhibition both in mothers (r = 0.818, p < 0.001) and infants (r = 0.386, p < 0.005). Previous SARS-CoV-2 infection and COVID-19 vaccination near delivery positively impacted anti-SARS-CoV-2 S IgG levels. CONCLUSIONS COVID-19 mRNA vaccines induce high anti-SARS-CoV-2 S titers in pregnant women, which can inhibit the binding of ACE2 to protein S and are efficiently transferred to the fetus. However, there was a rapid decrease in antibody levels at 2 to 3 months post-partum, particularly in infants.
Collapse
Affiliation(s)
- María José Muñoz-Gómez
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - María Martin-Vicente
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Sara Vigil-Vazquez
- Sevicio de Neonatología, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Itziar Carrasco
- Sevicio de Neonatología, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Alicia Hernanz Lobo
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Grupo de Investigación en Infectología Pediátrica, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Servicio de Pediatría, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Vicente Mas
- Unidad de Biología Viral, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - Mónica Vázquez
- Unidad de Biología Viral, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - Angela Manzanares
- Servicio de Pediatría, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Olga Cano
- Unidad de Biología Viral, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - Clara Zamora
- Servicio de Obstetricia y Ginecología, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Roberto Alonso
- Departamento de Microbiología Clínica y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Daniel Sepulveda-Crespo
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Tarancon-Diez
- Laboratorio de InmunoBiología Molecular, Sección de Inmunología. Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - María Ángeles Muñoz-Fernández
- Laboratorio de InmunoBiología Molecular, Sección de Inmunología. Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Spanish HIV-HGM BioBank, Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Mar Muñoz-Chapuli
- Servicio de Obstetricia y Ginecología, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Salvador Resino
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain.
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
| | - Maria Luisa Navarro
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Grupo de Investigación en Infectología Pediátrica, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Servicio de Pediatría, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Universidad Complutense de Madrid, Madrid, Spain
| | - Isidoro Martinez
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain.
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
5
|
Sinha D, Yaugel-Novoa M, Waeckel L, Paul S, Longet S. Unmasking the potential of secretory IgA and its pivotal role in protection from respiratory viruses. Antiviral Res 2024; 223:105823. [PMID: 38331200 DOI: 10.1016/j.antiviral.2024.105823] [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: 12/11/2023] [Revised: 01/22/2024] [Accepted: 01/25/2024] [Indexed: 02/10/2024]
Abstract
Mucosal immunity has regained its spotlight amidst the ongoing Coronavirus disease 19 (COVID-19) pandemic, with numerous studies highlighting the crucial role of mucosal secretory IgA (SIgA) in protection against Severe acute respiratory syndrome coronavirus-2 or SARS-CoV-2 infections. The observed limitations in the efficacy of currently authorized COVID-19 vaccines in inducing effective mucosal immune responses remind us of the limitations of systemic vaccination in promoting protective mucosal immunity. This resurgence of interest has motivated the development of vaccine platforms capable of enhancing mucosal responses, specifically the SIgA response, and the development of IgA-based therapeutics. Recognizing viral respiratory infections as a global threat, we would like to comprehensively review the existing knowledge on mucosal immunity, with a particular emphasis on SIgA, in the context of SARS-CoV-2, influenza, and Respiratory Syncytial Virus (RSV) infections. This review aims to describe the structural and functional specificities of SIgA, along with its nuanced role in combating influenza, RSV, and SARS-CoV-2 infections. Subsequent sections further elaborate promising vaccine strategies, including mucosal vaccines against Influenza, RSV, and SARS-CoV-2 respiratory viruses, currently undergoing preclinical and clinical development. Additionally, we address the challenges associated with mucosal vaccine development, concluding with a discussion on IgA-based therapeutics as a promising platform for the treatment of viral respiratory infections. This comprehensive review not only synthesizes current insights into mucosal immunity but also identifies critical knowledge gaps, strengthening the way for further advancements in our current understanding and approaches to combat respiratory viral threats.
Collapse
Affiliation(s)
- Divya Sinha
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, CIC 1408 Vaccinology, F42023, Saint-Etienne, France
| | - Melyssa Yaugel-Novoa
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, CIC 1408 Vaccinology, F42023, Saint-Etienne, France
| | - Louis Waeckel
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, CIC 1408 Vaccinology, F42023, Saint-Etienne, France; Immunology Department, University Hospital of Saint-Etienne, F42055, Saint-Etienne, France
| | - Stéphane Paul
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, CIC 1408 Vaccinology, F42023, Saint-Etienne, France; Immunology Department, University Hospital of Saint-Etienne, F42055, Saint-Etienne, France; CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, F42055, Saint-Etienne, France.
| | - Stéphanie Longet
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, CIC 1408 Vaccinology, F42023, Saint-Etienne, France.
| |
Collapse
|
6
|
Valverde-Merino MI, Gomez-Guzman M, Piquer-Martinez C, Cabezas Lopez MD, Zarzuelo MJ. The importance of COVID-19 vaccination during lactation. Infect Dis Now 2024; 54:104831. [PMID: 37952583 DOI: 10.1016/j.idnow.2023.104831] [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: 06/15/2023] [Revised: 10/24/2023] [Accepted: 11/03/2023] [Indexed: 11/14/2023]
Abstract
The World Health Organization strongly supports breastfeeding as the main source of infant feeding to ensure maternal and child health. Since its emergence, COVID-19 has become a disease affecting the health of the world's population, and vaccines have been developed to prevent it. However, the decision to license COVID-19 vaccines for infants under 6 months of age has been delayed. Different studies have shown that during the breastfeeding period, the benefit-risk balance is much higher in favor of the benefit, at the immunological level for the infant, due to its low perception of adverse effects and the low transmission of products such as mRNA from the mother to the child. Different organizations and societies recommend vaccination in breastfeeding women. COVID-19 vaccines have been shown to be safe and effective.
Collapse
Affiliation(s)
- Maria Isabel Valverde-Merino
- Pharmaceutical Care Research Group. Department of Pharmacy and Pharmaceutical Technology. Faculty of Pharmacy. University of Granada, Spain
| | - Manuel Gomez-Guzman
- Department of Pharmacology. Faculty of Pharmacy. University of Granada, Spain
| | - Celia Piquer-Martinez
- Pharmaceutical Care Research Group. Department of Pharmacy and Pharmaceutical Technology. Faculty of Pharmacy. University of Granada, Spain
| | - Maria Dolores Cabezas Lopez
- Pharmaceutical Care Research Group. Department of Pharmacy and Pharmaceutical Technology. Faculty of Pharmacy. University of Granada, Spain
| | - Maria Jose Zarzuelo
- Pharmaceutical Care Research Group. Department of Pharmacy and Pharmaceutical Technology. Faculty of Pharmacy. University of Granada, Spain.
| |
Collapse
|
7
|
Fernández-Buhigas I, Rayo N, Silos JC, Serrano B, Ocón-Hernández O, Leung BW, Delgado JL, Fernández DSN, Valle S, De Miguel L, Silgado A, Tanoira RP, Rolle V, Santacruz B, Gil MM, Poon LC. Anti-SARS-CoV-2-specific antibodies in human breast milk following SARS-CoV-2 infection during pregnancy: a prospective cohort study. Int Breastfeed J 2024; 19:5. [PMID: 38238855 PMCID: PMC10797875 DOI: 10.1186/s13006-023-00605-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 12/02/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND While the presence of SARS-CoV-2 in human breast milk is contentious, anti-SARS-CoV-2 antibodies have been consistently detected in human breast milk. However, it is uncertain when and how long the antibodies are present. METHODS This was a prospective cohort study including all consecutive pregnant women with confirmed SARS-CoV-2 infection during pregnancy, recruited at six maternity units in Spain and Hong Kong from March 2020 to March 2021. Colostrum (day of birth until day 4 postpartum) and mature milk (day 7 postpartum until 6 weeks postpartum) were prospectively collected, and paired maternal blood samples were also collected. Colostrum samples were tested with rRT-PCR-SARS-CoV-2, and skimmed acellular milk and maternal sera were tested against SARS-CoV-2 specific immunoglobulin M, A, and G reactive to receptor binding domain of SARS-CoV-2 spike protein 1 to determine the presence of immunoglobulins. Then, we examined how each immunoglobulin type in the colostrum was related to the time of infection by logistic regression analysis, the concordance between these immunoglobulins in the colostrum, maternal serum, and mature milk by Cohen's kappa statistic, and the relationship between immunoglobulin levels in mature milk and colostrum with McNemar. RESULTS One hundred eighty-seven pregnant women with confirmed SARS-CoV-2 infection during pregnancy or childbirth were recruited and donated the milk and blood samples. No SARS-CoV-2 was found in the human breast milk. Immunoglobulin A, G, and M were present in 129/162 (79·6%), 5/163 (3·1%), and 15/76 (19·7%) colostrum samples and in 17/62 (27·42%), 2/62 (3·23%) and 2/62 (3·23%) mature milk samples, respectively. Immunoglobulin A was the predominant immunoglobulin found in breast milk, and its levels were significantly higher in the colostrum than in the mature milk (p-value < 0.001). We did not find that the presence of immunoglobulins in the colostrum was associated with their presence in maternal, the severity of the disease, or the time when the infection had occurred. CONCLUSIONS Since anti-SARS-CoV-2 antibodies are found in the colostrum irrespective of the time of infection during pregnancy, but the virus itself is not detected in human breast milk, our study found no indications to withhold breastfeeding, taking contact precautions when there is active disease.
Collapse
Affiliation(s)
- Irene Fernández-Buhigas
- Obstetrics and Gynecology Department, Hospital Universitario de Torrejón, Torrejón de Ardoz, Madrid, Spain
- Vicerrectorado de Investigación, Facultad de Medicina, Universidad Francisco de Vitoria, Carretera Pozuelo a Majadahonda, Km 1.800, Pozuelo de Alarcón, Madrid, 28223, Spain
| | - Nieves Rayo
- Obstetrics and Gynecology Department, Hospital Universitario de Torrejón, Torrejón de Ardoz, Madrid, Spain
- Vicerrectorado de Investigación, Facultad de Medicina, Universidad Francisco de Vitoria, Carretera Pozuelo a Majadahonda, Km 1.800, Pozuelo de Alarcón, Madrid, 28223, Spain
| | - Julia Cuesta Silos
- Synlab Diagnósticos Globales S.A., Esplugues de Llobregat, Catalonia, Spain
| | - Berta Serrano
- Department of Obstetrics, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Catalonia, Spain
| | - Olga Ocón-Hernández
- Obstetrics and Gynecology Department, Hospital Universitario Clínico San Cecilio, Granada, Spain
- Instituto de Investigación Biosanitaria Ibs, Granada, Spain
| | - Bo Wah Leung
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Juan Luis Delgado
- Obstetrics and Gynecology Department, Hospital Clínico Universitario Virgen de La Arrixaca, El Palmar, Murcia, Spain
| | - David Sánchez-Nieves Fernández
- Obstetrics and Gynecology Department, Hospital Universitário Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
- Universidad de Alcalá de Henares, School of Medicine, Alcalá de Henares, Madrid, Spain
| | - Silvia Valle
- Obstetrics and Gynecology Department, Hospital Universitario de Torrejón, Torrejón de Ardoz, Madrid, Spain
- Vicerrectorado de Investigación, Facultad de Medicina, Universidad Francisco de Vitoria, Carretera Pozuelo a Majadahonda, Km 1.800, Pozuelo de Alarcón, Madrid, 28223, Spain
| | - Laura De Miguel
- Synlab Diagnósticos Globales S.A., Esplugues de Llobregat, Catalonia, Spain
| | - Aroa Silgado
- Department of Microbiology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Catalonia, Spain
| | - Ramón Perez Tanoira
- Department of Microbiology, Hospital Universitário Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Valeria Rolle
- Biostatistics and Epidemiology Platform at Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Asturias, Spain
| | - Belén Santacruz
- Obstetrics and Gynecology Department, Hospital Universitario de Torrejón, Torrejón de Ardoz, Madrid, Spain
- Vicerrectorado de Investigación, Facultad de Medicina, Universidad Francisco de Vitoria, Carretera Pozuelo a Majadahonda, Km 1.800, Pozuelo de Alarcón, Madrid, 28223, Spain
| | - Maria M Gil
- Obstetrics and Gynecology Department, Hospital Universitario de Torrejón, Torrejón de Ardoz, Madrid, Spain.
- Vicerrectorado de Investigación, Facultad de Medicina, Universidad Francisco de Vitoria, Carretera Pozuelo a Majadahonda, Km 1.800, Pozuelo de Alarcón, Madrid, 28223, Spain.
| | - Liona C Poon
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
8
|
de Graaf SC, Bondt A, van Rijswijck DMH, Juncker HG, Mulleners SJ, Damen MJA, Hoek M, van Keulen BJ, van Goudoever JB, Heck AJR, Dingess KA. A case series exploring the human milk polyclonal IgA1 response to repeated SARS-CoV-2 vaccinations by LC-MS based fab profiling. Front Nutr 2024; 10:1305086. [PMID: 38288064 PMCID: PMC10822949 DOI: 10.3389/fnut.2023.1305086] [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: 10/03/2023] [Accepted: 12/20/2023] [Indexed: 01/31/2024] Open
Abstract
Introduction Upon vaccination against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) humans will start to produce antibodies targeting virus specific antigens that will end up in circulation. In lactating women such antibodies will also end up in breastmilk, primarily in the form of secretory immunoglobulin A1 (SIgA1), the most abundant immunoglobulin (Ig) in human milk. Here we set out to investigate the SIgA1 clonal repertoire response to repeated SARS-CoV-2 vaccination, using a LC-MS fragment antigen-binding (Fab) clonal profiling approach. Methods We analyzed the breastmilk of six donors from a larger cohort of 109 lactating mothers who received one of three commonly used SARS-CoV-2 vaccines. We quantitatively monitored the SIgA1 Fab clonal profile over 16 timepoints, from just prior to the first vaccination until 15 days after the second vaccination. Results In all donors, we detected a population of 89-191 vaccine induced clones. These populations were unique to each donor and heterogeneous with respect to individual clonal concentrations, total clonal titer, and population size. The vaccine induced clones were dominated by persistent clones (68%) which came up after the first vaccination and were retained or reoccurred after the second vaccination. However, we also observe transient SIgA1 clones (16%) which dissipated before the second vaccination, and vaccine induced clones which uniquely emerged only after the second vaccination (16%). These distinct populations were observed in all analyzed donors, regardless of the administered vaccine. Discussion Our findings suggest that while individual donors have highly unique human milk SIgA1 clonal profiles and a highly personalized SIgA1 response to SARS-CoV-2 vaccination, there are also commonalities in vaccine induced responses.
Collapse
Affiliation(s)
- Sebastiaan C. de Graaf
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Albert Bondt
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Danique M. H. van Rijswijck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Hannah G. Juncker
- Department of Pediatrics, Amsterdam Reproduction and Development Research Institute, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, Netherlands
| | - Sien J. Mulleners
- Department of Pediatrics, Amsterdam Reproduction and Development Research Institute, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Mirjam J. A. Damen
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Max Hoek
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Britt J. van Keulen
- Department of Pediatrics, Amsterdam Reproduction and Development Research Institute, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Johannes B. van Goudoever
- Department of Pediatrics, Amsterdam Reproduction and Development Research Institute, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Albert J. R. Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Kelly A. Dingess
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
- Department of Pediatrics, Amsterdam Reproduction and Development Research Institute, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
9
|
Miyamoto S, Nishiyama T, Ueno A, Park H, Kanno T, Nakamura N, Ozono S, Aihara K, Takahashi K, Tsuchihashi Y, Ishikane M, Arashiro T, Saito S, Ainai A, Hirata Y, Iida S, Katano H, Tobiume M, Tokunaga K, Fujimoto T, Suzuki M, Nagashima M, Nakagawa H, Narita M, Kato Y, Igari H, Fujita K, Kato T, Hiyama K, Shindou K, Adachi T, Fukushima K, Nakamura-Uchiyama F, Hase R, Yoshimura Y, Yamato M, Nozaki Y, Ohmagari N, Suzuki M, Saito T, Iwami S, Suzuki T. Infectious virus shedding duration reflects secretory IgA antibody response latency after SARS-CoV-2 infection. Proc Natl Acad Sci U S A 2023; 120:e2314808120. [PMID: 38134196 PMCID: PMC10756199 DOI: 10.1073/pnas.2314808120] [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: 08/26/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
Infectious virus shedding from individuals infected with severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) is used to estimate human-to-human transmission risk. Control of SARS-CoV-2 transmission requires identifying the immune correlates that protect infectious virus shedding. Mucosal immunity prevents infection by SARS-CoV-2, which replicates in the respiratory epithelium and spreads rapidly to other hosts. However, whether mucosal immunity prevents the shedding of the infectious virus in SARS-CoV-2-infected individuals is unknown. We examined the relationship between viral RNA shedding dynamics, duration of infectious virus shedding, and mucosal antibody responses during SARS-CoV-2 infection. Anti-spike secretory IgA antibodies (S-IgA) reduced viral RNA load and infectivity more than anti-spike IgG/IgA antibodies in infected nasopharyngeal samples. Compared with the IgG/IgA response, the anti-spike S-IgA post-infection responses affected the viral RNA shedding dynamics and predicted the duration of infectious virus shedding regardless of the immune history. These findings highlight the importance of anti-spike S-IgA responses in individuals infected with SARS-CoV-2 for preventing infectious virus shedding and SARS-CoV-2 transmission. Developing medical countermeasures to shorten S-IgA response time may help control human-to-human transmission of SARS-CoV-2 infection and prevent future respiratory virus pandemics.
Collapse
Affiliation(s)
- Sho Miyamoto
- Department of Pathology, National Institute of Infectious Diseases, Tokyo162-8640, Japan
| | - Takara Nishiyama
- Interdisciplinary Biology Laboratory, Division of Natural Science, Graduate School of Science, Nagoya University, Aichi464-8602, Japan
| | - Akira Ueno
- Department of Pathology, National Institute of Infectious Diseases, Tokyo162-8640, Japan
| | - Hyeongki Park
- Interdisciplinary Biology Laboratory, Division of Natural Science, Graduate School of Science, Nagoya University, Aichi464-8602, Japan
| | - Takayuki Kanno
- Department of Pathology, National Institute of Infectious Diseases, Tokyo162-8640, Japan
| | - Naotoshi Nakamura
- Interdisciplinary Biology Laboratory, Division of Natural Science, Graduate School of Science, Nagoya University, Aichi464-8602, Japan
| | - Seiya Ozono
- Department of Pathology, National Institute of Infectious Diseases, Tokyo162-8640, Japan
| | - Kazuyuki Aihara
- International Research Center for Neurointelligence, The University of Tokyo Institutes for Advanced Study, The University of Tokyo, Tokyo113-0033, Japan
| | - Kenichiro Takahashi
- Center for Emergency Preparedness and Response, National Institute of Infectious Diseases, Tokyo162-8640, Japan
| | - Yuuki Tsuchihashi
- Center for surveillance, Immunization, and Epidemiologic Research, National Institute of Infectious Diseases, Tokyo162-8640, Japan
- Center for Field Epidemic Intelligence, Research and Professional Development, National Institute of Infectious Diseases, Tokyo162-8640, Japan
| | - Masahiro Ishikane
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo162-8655, Japan
| | - Takeshi Arashiro
- Department of Pathology, National Institute of Infectious Diseases, Tokyo162-8640, Japan
- Center for surveillance, Immunization, and Epidemiologic Research, National Institute of Infectious Diseases, Tokyo162-8640, Japan
| | - Shinji Saito
- Department of Pathology, National Institute of Infectious Diseases, Tokyo162-8640, Japan
| | - Akira Ainai
- Department of Pathology, National Institute of Infectious Diseases, Tokyo162-8640, Japan
| | - Yuichiro Hirata
- Department of Pathology, National Institute of Infectious Diseases, Tokyo162-8640, Japan
| | - Shun Iida
- Department of Pathology, National Institute of Infectious Diseases, Tokyo162-8640, Japan
| | - Harutaka Katano
- Department of Pathology, National Institute of Infectious Diseases, Tokyo162-8640, Japan
| | - Minoru Tobiume
- Department of Pathology, National Institute of Infectious Diseases, Tokyo162-8640, Japan
| | - Kenzo Tokunaga
- Department of Pathology, National Institute of Infectious Diseases, Tokyo162-8640, Japan
| | - Tsuguto Fujimoto
- Center for Emergency Preparedness and Response, National Institute of Infectious Diseases, Tokyo162-8640, Japan
| | - Michiyo Suzuki
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo162-8655, Japan
| | - Maki Nagashima
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo162-8655, Japan
| | - Hidenori Nakagawa
- Department of Infectious Diseases, Osaka City General Hospital, Osaka534-0021, Japan
| | - Masashi Narita
- Division of Infectious Diseases, Department of Internal Medicine, Okinawa Prefectural Nanbu Medical Center and Children’s Medical Center, Okinawa901-1193, Japan
| | - Yasuyuki Kato
- Department of Infectious Diseases, International University of Health and Welfare Narita Hospital, Chiba286-0124, Japan
| | - Hidetoshi Igari
- Department of Infection Control, Chiba University Hospital, Chiba, Japan
| | - Kaori Fujita
- Department of Respiratory Medicine, National Hospital Organization Okinawa National Hospital, Okinawa901-2214, Japan
| | - Tatsuo Kato
- Department of Chest Disease, National Hospital Organization Nagara Medical Center, Gifu502-8558, Japan
| | - Kazutoshi Hiyama
- Department of Infectious Disease, National Hospital Organization Fukuoka-Higashi Medical Center, Fukuoka811-3195, Japan
| | - Keisuke Shindou
- Department of Pediatrics, Hirakata City Hospital, Osaka573-1013, Japan
| | - Takuya Adachi
- Department of Infectious Diseases, Tokyo Metropolitan Toshima Hospital, Tokyo173-0015, Japan
| | - Kazuaki Fukushima
- Department of Infectious Disease, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo113-8677, Japan
| | | | - Ryota Hase
- Department of Infectious Diseases, Japanese Red Cross Narita Hospital, Chiba286-8523, Japan
| | - Yukihiro Yoshimura
- Division of Infectious Disease, Yokohama Municipal Citizen’s Hospital, Kanagawa221-0855, Japan
| | - Masaya Yamato
- Department of General Internal Medicine and Infectious Diseases, Rinku General Medical Center 598-8577, Osaka, Japan
| | - Yasuhiro Nozaki
- Department of Respiratory Medicine, Tokoname City Hospital, Aichi479-8510, Japan
| | - Norio Ohmagari
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo162-8655, Japan
| | - Motoi Suzuki
- Center for surveillance, Immunization, and Epidemiologic Research, National Institute of Infectious Diseases, Tokyo162-8640, Japan
| | - Tomoya Saito
- Center for Emergency Preparedness and Response, National Institute of Infectious Diseases, Tokyo162-8640, Japan
| | - Shingo Iwami
- Interdisciplinary Biology Laboratory, Division of Natural Science, Graduate School of Science, Nagoya University, Aichi464-8602, Japan
- Institute of Mathematics for Industry, Kyushu University, Fukuoka819-0395, Japan
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto606-8501, Japan
- Interdisciplinary Theoretical and Mathematical Sciences Program, RIKEN, Saitama351-0198, Japan
- NEXT-Ganken Program, Japanese Foundation for Cancer Research, Tokyo135-8550, Japan
- Science Groove Inc., Fukuoka810-0041, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo162-8640, Japan
| |
Collapse
|
10
|
Leontieva G, Gupalova T, Desheva Y, Kramskaya T, Bormotova E, Koroleva I, Kopteva O, Suvorov A. Evaluation of Immune Response to Mucosal Immunization with an Oral Probiotic-Based Vaccine in Mice: Potential for Prime-Boost Immunization against SARS-CoV-2. Int J Mol Sci 2023; 25:215. [PMID: 38203387 PMCID: PMC10779021 DOI: 10.3390/ijms25010215] [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: 10/31/2023] [Revised: 12/06/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Following the conclusion of the COVID-19 pandemic, the persistent genetic variability in the virus and its ongoing circulation within the global population necessitate the enhancement of existing preventive vaccines and the development of novel ones. A while back, we engineered an orally administered probiotic-based vaccine, L3-SARS, by integrating a gene fragment that encodes the spike protein S of the SARS-CoV-2 virus into the genome of the probiotic strain E. faecium L3, inducing the expression of viral antigen on the surface of bacteria. Previous studies demonstrated the efficacy of this vaccine candidate in providing protection against the virus in Syrian hamsters. In this present study, utilizing laboratory mice, we assess the immune response subsequent to immunization via the gastrointestinal mucosa and discuss its potential as an initial phase in a two-stage vaccination strategy. Our findings indicate that the oral administration of L3-SARS elicits an adaptive immune response in mice. Pre-immunization with L3-SARS enhances and prolongs the humoral immune response following a single subcutaneous immunization with a recombinant S-protein analogous to the S-insert of the coronavirus in Enterococcus faecium L3.
Collapse
Affiliation(s)
| | | | - Yulia Desheva
- Scientific and Educational Center, Molecular Bases of Interaction of Microorganisms and Human of the World-Class Research Center, Center for Personalized Medicine, FSBSI, IEM, 197376 Saint Petersburg, Russia; (G.L.); (T.G.); (T.K.); (E.B.); (I.K.); (O.K.); (A.S.)
| | | | | | | | | | | |
Collapse
|
11
|
Vaddi VK, Mascarenhas D, Kirthana SB, Mundhra N, Nanavati R. A Quality Improvement Initiative to Increase the Milk Donation to the Human Milk Bank Post-Coronavirus Disease-19 Pandemic. Breastfeed Med 2023; 18:864-869. [PMID: 37733277 DOI: 10.1089/bfm.2023.0124] [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] [Indexed: 09/22/2023]
Abstract
Background and Objective: Donor human milk (DHM) from the human milk bank (HMB) is the next best alterative in circumstances when mother's own milk is not available. There was a steep decline in the volume of DHM collected during the coronavirus disease-19 (COVID-19) pandemic due to various factors, while DHM demand increased. Hence, a quality improvement (QI) study was conducted to increase the volume of milk donation to HMB from postpandemic baseline of 300-400 to 1,000 mL/day over 8 weeks. Materials and Methods: Fish bone analysis was used to identify the potential barriers, and four Plan-Do-Study-Act (PDSA) cycles were conducted from January 2021 to March 2021 to address the key barriers. In the first PDSA cycle, training of health care providers was done. Sessions for educating mothers in the second PDSA cycle and individualized one-to-one counseling of mothers by a mother support group were done in the third PDSA cycle. The availability of breast pump was increased in the fourth PDSA cycle. Sustainability of the interventions was studied for 6 months and data were analyzed. Results: The average DHM collected per day at the end of each PDSA cycle was 900, 1,500, 1,000, and 1,100 mL. Although the sustenance phase was affected by the second COVID-19 wave, prompt identification of the issues and timely interventions prevented the donated volume from dropping to preintervention levels. Conclusion: QI initiatives customized for local settings can result in significant improvement in voluntary milk donation in HMB, which can result in more availability of DHM to premature babies.
Collapse
Affiliation(s)
- Vamsi Krishna Vaddi
- Department of Neonatology, Seth GS Medical College and KEM Hospital, Mumbai, India
| | - Dwayne Mascarenhas
- Division of Neonatology, The Hospital for Sick Children, Toronto, Canada
| | - S B Kirthana
- Department of Neonatology, Seth GS Medical College and KEM Hospital, Mumbai, India
| | - Nitu Mundhra
- Department of Neonatology, Wockhardt Hospital, Mumbai, India
| | - Ruchi Nanavati
- Department of Neonatology, Seth GS Medical College and KEM Hospital, Mumbai, India
| |
Collapse
|
12
|
Zöllkau J, Heimann Y, Hagenbeck C, Pecks U, Abou-Dakn M, Schlösser R, Schohe A, Dressler-Steinbach I, Manz M, Banz-Jansen C, Reuschel E, Iannaccone A, Bohlmann MK, Kraft K, Fill Malfertheiner S, Wimberger P, Kolben T, Bartmann C, Longardt AC. Breastfeeding Behavior Within the Covid-19 Related Obstetric and Neonatal Outcome Study (CRONOS). J Hum Lact 2023; 39:625-635. [PMID: 37712573 DOI: 10.1177/08903344231190623] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
BACKGROUND The SARS-CoV-2 pandemic and its influence on peripartum processes worldwide led to issues in breastfeeding support. RESEARCH AIM The aim of this study was to describe breastfeeding behavior and peripartum in-hospital management during the pandemic in Germany and Austria. METHODS This study was a descriptive study using a combination of secondary longitudinal data and a cross-sectional online survey. Registry data from the prospective multicenter COVID-19 Related Obstetric and Neonatal Outcome Study (CRONOS) cohort study (longitudinal, medical records of 1,815 parent-neonate pairs with confirmed SARS-CoV-2 infection during pregnancy) and a cross-sectional online survey of CRONOS hospitals' physicians (N = 67) were used for a descriptive comparison of feeding outcomes and postpartum management. RESULTS In 93.7% (n = 1700) of the cases in which information on the neonate's diet was provided, feeding was with the mother's own milk. Among neonates not receiving their mother's own milk, 24.3% (n = 26) reported SARS-CoV-2 infection as the reason. Peripartum maternal SARS-CoV-2 infection, severe maternal COVID-19 including the need for intensive care unit (ICU) treatment or invasive ventilation, preterm birth, mandatory delivery due to COVID-19, and neonatal ICU admission were associated with lower rates of breastfeeding. Rooming-in positively influenced breastfeeding without affecting neonatal SARS-CoV-2 frequency (4.2% vs. 5.6%). CRONOS hospitals reported that feeding an infant their mother's own milk continued to be supported during the pandemic. In cases of severe COVID-19, four of five hospitals encouraged breastfeeding. CONCLUSION Maintaining rooming-in and breastfeeding support services in the CRONOS hospitals during the pandemic resulted in high breastfeeding rates.
Collapse
Affiliation(s)
- Janine Zöllkau
- Department of Obstetrics, Jena University Hospital, Jena, Germany
| | - Yvonne Heimann
- Department of Obstetrics, Jena University Hospital, Jena, Germany
| | - Carsten Hagenbeck
- Clinic for Gynecology and Obstetrics, Heinrich-Heine-University, Düsseldorf, Germany
| | - Ulrich Pecks
- Department of Obstetrics and Gynecology, University Hospital of Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Michael Abou-Dakn
- Department of Obstetrics and Gynecology, St. Joseph-Hospital, Berlin, Germany
| | - Rolf Schlösser
- Hospital for Children and Adolescents, University Hospital of Frankfurt, Frankfurt, Germany
| | - Anna Schohe
- Department of Obstetrics and Gynecology, St. Joseph-Hospital, Berlin, Germany
| | | | - Maike Manz
- Department of Obstetrics and Gynecology, Darmstadt City Hospital, Darmstadt, Germany
| | - Constanze Banz-Jansen
- Department of Gynecology and Obstetrics, Protestant Hospital of Bethel Foundation, Bielefeld, Germany
| | - Edith Reuschel
- University Department of Obstetrics and Gynecology, The Hospital St. Hedwig of The Order of St. John, University of Regensburg, Regensburg, Germany
| | | | - Michael K Bohlmann
- Department of Obstetrics and Gynecology, St. Elisabeth' Hospital, Loerrach, Germany
| | - Katrina Kraft
- Department of Obstetrics and Gynecology, Munich Municipal Hospital, Harlaching, Munich, Germany
| | - Sara Fill Malfertheiner
- University Department of Obstetrics and Gynecology, The Hospital St. Hedwig of The Order of St. John, University of Regensburg, Regensburg, Germany
| | - Pauline Wimberger
- Department of Gynecology and Obstetrics, TU Dresden, Dresden, Germany
| | - Thomas Kolben
- Department for Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Catharina Bartmann
- Department of Gynecology and Obstetrics, University Hospital Würzburg, Würzburg, Germany
| | - Ann - Carolin Longardt
- Clinic for Pediatrics and Adolescent Medicine I / Neonatology, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| |
Collapse
|
13
|
Golan Y, Ilala M, Li L, Gay C, Hunagund S, Lin CY, Cassidy AG, Jigmeddagva U, Matsui Y, Ozarslan N, Asiodu IV, Ahituv N, Flaherman VJ, Gaw SL, Prahl M. Milk antibody response after 3 rd COVID-19 vaccine and SARS-CoV-2 infection and implications for infant protection. iScience 2023; 26:107767. [PMID: 37731614 PMCID: PMC10507209 DOI: 10.1016/j.isci.2023.107767] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 07/28/2023] [Accepted: 08/25/2023] [Indexed: 09/22/2023] Open
Abstract
Little is known about the persistence of human milk anti-SARS-CoV-2 antibodies after 2nd and 3rd vaccine doses and infection following 3rd dose. In this study, human milk, saliva, and blood samples were collected from 33 lactating individuals before and after vaccination and infection. Antibody levels were measured using ELISA and symptoms were assessed using questionnaires. We found that after vaccination, milk anti-SARS-CoV-2 antibodies persisted for up to 8 months. In addition, distinct patterns of human milk IgA and IgG production and higher milk RBD-blocking activity was observed after infection compared to 3-dose vaccination. Infected mothers reported more symptoms than vaccinated mothers. We examined the persistence of milk antibodies in infant saliva after breastfeeding and found that IgA was more abundant compared to IgG. Our results emphasize the importance of improving the secretion of IgA antibodies to human milk after vaccination to improve the protection of breastfeeding infants.
Collapse
Affiliation(s)
- Yarden Golan
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Mikias Ilala
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Lin Li
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Caryl Gay
- Department of Family Health Care Nursing, University of California, San Francisco, San Francisco, CA, USA
| | - Soumya Hunagund
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and 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
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, 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
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Unurzul Jigmeddagva
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Yusuke Matsui
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, 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
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Ifeyinwa V. Asiodu
- Department of Family Health Care Nursing, University of California, San Francisco, San Francisco, CA, USA
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Valerie J. Flaherman
- Department of Pediatrics, 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
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Mary 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
| |
Collapse
|
14
|
Antony MA, Patel S, Verma V, Kant R. The Role of Gut Microbiome Supplementation in COVID-19 Management. Cureus 2023; 15:e46960. [PMID: 38021562 PMCID: PMC10640765 DOI: 10.7759/cureus.46960] [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] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
COVID-19, which is caused by the RNA virus, SARS-CoV-2, mainly affects the respiratory system and has a varied clinical presentation. However, several studies have shown that COVID-19 can also affect the gastrointestinal (GI) system. Patients can experience various GI symptoms, such as vomiting and diarrhea, and the virus has been detected in the stool samples of patients hospitalized with COVID-19. There have also been rare reports of COVID-19 presenting with isolated GI symptoms and lack of respiratory symptoms, and the virus has also been detected for prolonged periods in the fecal samples of COVID-19 patients. Major alterations in the gut microbiome in the form of depletion of beneficial organisms and an abundance of pathogenic organisms have been reported in the fecal samples of hospitalized COVID-19 patients. Although the US FDA has approved several drugs to manage COVID-19, their efficacy remains modest. So, there is a constant ongoing effort to investigate novel treatment options for COVID-19. Health supplements like probiotics, prebiotics, postbiotics, and synbiotics have been popularly known for their various health benefits. In this review, we have summarized the current literature, which shows the potential benefit of these health supplements to mitigate and/or prevent the clinical presentation of COVID-19.
Collapse
Affiliation(s)
- Mc Anto Antony
- Department of Endocrinology, Diabetes and Metabolism, Medical University of South Carolina, Anderson, USA
| | - Siddharth Patel
- Department of Internal Medicine, Decatur Morgan Hospital, Decatur, USA
| | - Vipin Verma
- Department of Internal Medicine, Medical University of South Carolina, Anderson, USA
| | - Ravi Kant
- Department of Endocrinology, Diabetes and Metabolism, Medical University of South Carolina, Anderson, USA
| |
Collapse
|
15
|
Morniroli D, Signorini L, Dolci M, Vizzari G, Ronchi A, Pietrasanta C, Pugni L, Mosca F, Delbue S, Gianni ML. Breastmilk from COVID-19 negative lactating mothers shows neutralizing activity against SARS-COV-2. Sci Rep 2023; 13:15521. [PMID: 37726309 PMCID: PMC10509262 DOI: 10.1038/s41598-023-42421-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 09/10/2023] [Indexed: 09/21/2023] Open
Abstract
Breastmilk protects newborns from infections through specific and nonspecific compounds. This study investigated the neutralizing activity against SARS-CoV-2 of breastmilk from SARS-CoV-2 negative, unvaccinated mothers, and compared it to that from infected nursing mothers. We enrolled women after COVID-19 swab testing results upon maternity admission, and divided them into two groups: group A, COVID-19-positive mothers, and group B, negative mothers. Breastmilk was randomly sampled at 2, 7, and 20 days postpartum. We collected 19 samples for Group A and 41 for Group B. A microneutralization assay was used to determine the 50% neutralization (NT50) titre. The presence of neutralizing antibodies was also determined. Group A had 100% neutralizing samples at 2 days postpartum (T0), declining 7 days postpartum (T1) and 20 days postpartum (T2). Group B samples exhibited neutralizing activity mostly at 7 days postpartum (T1) (90%). Negative mothers' samples showed no correlation between NT50 titres and antibodies' presence, suggesting that non-specific breastmilk components may exert antiviral action against SARS-CoV-2.
Collapse
Affiliation(s)
- Daniela Morniroli
- Department of Clinical Sciences and Community Health, University of Milan, Via Della Commenda 12, 20122, Milan, Italy.
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy.
| | - Lucia Signorini
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Maria Dolci
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Giulia Vizzari
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
| | - Andrea Ronchi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
| | - Carlo Pietrasanta
- Department of Clinical Sciences and Community Health, University of Milan, Via Della Commenda 12, 20122, Milan, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
| | - Lorenza Pugni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
| | - Fabio Mosca
- Department of Clinical Sciences and Community Health, University of Milan, Via Della Commenda 12, 20122, Milan, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
| | - Serena Delbue
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Maria Lorella Gianni
- Department of Clinical Sciences and Community Health, University of Milan, Via Della Commenda 12, 20122, Milan, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
| |
Collapse
|
16
|
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
|
17
|
He YF, Liu JQ, Hu XD, Li HM, Wu N, Wang J, Jiang ZG. Breastfeeding vs. breast milk transmission during COVID-19 pandemic, which is more important? Front Pediatr 2023; 11:1253333. [PMID: 37744448 PMCID: PMC10511770 DOI: 10.3389/fped.2023.1253333] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
The catastrophic coronavirus disease 2019 (COVID-19) pandemic has raised many health questions, and whether breast milk from SARS-CoV-2 infected mothers may be a vector for SARS-CoV-2 transmission has become a hot topic of concern worldwide. Currently, there are extremely limited and conflicting data on the risk of infection in infants through breastfeeding. For this reason, we investigated almost all current clinical studies and systematically analyzed the presence of SARS-CoV-2 and antibodies in the breast milk of mothers infected with SARS-CoV-2, their effects on newborns, and the mechanisms involved. A total of 82 studies were included in this review, of which 66 examined the presence of SARS-CoV-2 in breast milk samples from mothers diagnosed with COVID-19, 29 reported results of antibody detection of SARS-CoV-2 in breast milk, and 13 reported both nucleic acid and antibody test results. Seventeen studies indicated the presence of detectable SARS-CoV-2 nucleic acid in breast milk samples, and only two studies monitored viral activity, both of which reported that infectious viruses could not be cultured from RNA-positive breast milk samples. All 29 studies indicated the presence of at least one of the three antibodies, IgA, IgG and IgM, in breast milk. Five studies indicated the presence of at least one antibody in the serum of breastfed newborns. No COVID-19-related deaths were reported in all 1,346 newborns. Our study suggests that direct breastfeeding does not pose an additional risk of infection to newborns and that breast milk is a beneficial source of anti-SARS-CoV-2 antibodies that provide passive immune protection to infants. In addition, direct breastfeeding would provide maternal benefits. Our review supports the recommendation to encourage direct breastfeeding under appropriate infection control guidelines. Systematic Review Registration: https://www.crd.york.ac.uk/PROSPERO/#myprospero, identifier: 458043.
Collapse
Affiliation(s)
- Yan-fei He
- Health Management Center, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jun-qiang Liu
- Department of Thoracic Surgery, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiao-dong Hu
- Department of Endocrinology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hu-ming Li
- Department of Respiratory Medicine, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ni Wu
- Health Management Center, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jie Wang
- Health Management Center, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhi-gang Jiang
- Department of Statistics, Zunyi Medical University, Zunyi, China
| |
Collapse
|
18
|
Mane V, Mehta R, Alvarez N, Sharma V, Park S, Fox A, DeCarlo C, Yang X, Perlin DS, Powell RLR. In Vivo Antiviral Efficacy of LCTG-002, a Pooled, Purified Human Milk Secretory IgA product, Against SARS-CoV-2 in a Murine Model of COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.25.554813. [PMID: 37693438 PMCID: PMC10491103 DOI: 10.1101/2023.08.25.554813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Immunoglobulin A (IgA) is the most abundant antibody (Ab) in human mucosal compartments including the respiratory tract, with the secretory form of IgA (sIgA) being dominant and uniquely stable in these environments. sIgA is naturally found in human milk, which could be considered a global resource for this biologic, justifying the development of human milk sIgA as a dedicated airway therapeutic for respiratory infections such as SARS-CoV-2. In the present study, methods were therefore developed to efficiently extract human milk sIgA from donors who were either immunologically naïve to SARS-CoV-2 (pooled as a control IgA) or had recovered from a PCR-confirmed SARS-CoV-2 infection that elicited high-titer anti-SARS-CoV-2 Spike sIgA Abs in their milk (pooled together to make LCTG-002). Mass spectrometry determined that proteins with a relative abundance of 1.0% or greater were all associated with sIgA. None of the proteins exhibited statistically significant differences between batches. Western blot demonstrated all batches consisted predominantly of sIgA. Compared to control IgA, LCTG-002 demonstrated significantly higher binding to Spike, and was also capable of blocking the Spike - ACE2 interaction in vitro with 6.3x greater potency compared to control IgA (58% inhibition at ∼240ug/mL). LCTG-002 was then tested in vivo for its capacity to reduce viral burden in the lungs of K18+hACE2 transgenic mice inoculated with SARS-CoV-2. LCTG-002 was demonstrated to significantly reduce SARS-CoV-2 titers in the lungs compared to control IgA when administered at either 250ug/day or 1 mg/day, as measured by TCID50, plaque forming units (PFU), and qRT-PCR, with a maximum reduction of 4.9 logs. This innovative study demonstrates that LCTG-002 is highly pure, efficacious, and well tolerated in vivo, supporting further development of milk-derived, polyclonal sIgA therapeutics against SARS-CoV-2 and other mucosal infections.
Collapse
|
19
|
Juncker HG, van Doesburg M, de Groot CJ, Pajkrt D, Korosi A, van Gils MJ, van Goudoever JB, van Keulen BJ. Physical activity in lactating women influences SARS-CoV-2-specific antibodies in human milk. Heliyon 2023; 9:e19218. [PMID: 37654447 PMCID: PMC10466918 DOI: 10.1016/j.heliyon.2023.e19218] [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: 09/23/2022] [Revised: 06/27/2023] [Accepted: 08/16/2023] [Indexed: 09/02/2023] Open
Abstract
Background Moderate exercise results in a significant increase in serum and salivary immunoglobulins. Maternal physical activity might therefore also be a factor influencing antibody levels in human milk. This study aims to determine the influence of physical activity on SARS-CoV-2-specific Immunoglobulin A (IgA) in human milk and Immunoglobulin G (IgG) in serum. Methods In this prospective cross-sectional cohort study, all lactating women in the Netherlands were eligible to participate. SARS-CoV-2-specific IgA in human milk and IgG in serum were determined using an enzyme-linked immunosorbent assay (ELISA). Data on performed physical activity was collected using the Short Questionnaire to Assess Health enhancing physical activity (SQUASH), which includes intensity and duration of the performed activity. Findings In total, 356 out of 2312 lactating women tested positive for SARS-CoV-2-specific antibodies in serum. Of them, 323 filled in the questionnaire and were included in the analysis. An association between the activity score and SARS-CoV-2-specific antibodies in human milk (B = 1·035, 95·0% CI = 1·019 to 1·052, p = 0·042) and serum (B = 1·019, 95·0% CI = 1·009 to 1·029, p = 0·048) was demonstrated. No association was found between the duration of physical activity and SARS-CoV-2-specific antibodies in human milk or serum. Interpretation Our findings suggest that physical activity is beneficial for the levels of SARS-CoV-2-specific antibodies in human milk and serum, with the intensity of the physical activity being the most important contributor to this relationship. A higher level of antibodies in human milk might provide better immunological protection for infants against COVID-19.
Collapse
Affiliation(s)
- Hannah G. Juncker
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
- Swammerdam Institute for Life Sciences - Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | - Maritt van Doesburg
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
| | - Christianne J.M. de Groot
- Amsterdam UMC, Vrije Universiteit, Amsterdam Reproduction & Development Research Institute, Department of Obstetrics and Gynaecology, Amsterdam, the Netherlands
| | - Dasja Pajkrt
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
| | - Aniko Korosi
- Swammerdam Institute for Life Sciences - Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | - Marit J. van Gils
- Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Department of Medical Microbiology and Infection Prevention, Amsterdam, the Netherlands
| | - Johannes B. van Goudoever
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
| | - Britt J. van Keulen
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
| |
Collapse
|
20
|
Karimi H, Mansouri V, Rezaei N. Vertical transmission and maternal passive immunity post-SARS-CoV-2. Future Virol 2023:10.2217/fvl-2023-0089. [PMID: 37822684 PMCID: PMC10564388 DOI: 10.2217/fvl-2023-0089] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/22/2023] [Indexed: 10/13/2023]
Abstract
Since 2020, the highly contagious nature and various transmission routes of SARS-CoV-2 have rendered the pandemic interminable. Vertical transmission (VT) through the placenta and breast milk, which is frequent for certain virus types, is thought to exist for SARS-CoV-2 and is hypothesized by many researchers. Conversely, antibodies are produced to counteract the effect of viruses. Since newborns' immunologic system cannot produce proper antibodies, maternal antibodies are usually transferred from mother to infant/fetus to meet the need. This theory leads to the hypothesis of transmission of antibodies through the placenta and breast milk following SARS-CoV-2 infection or vaccination. This paper further discusses these hypotheses, considering consequences of fetus/infant harm versus benefit.
Collapse
Affiliation(s)
- Hanie Karimi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Vahid Mansouri
- Gene Therapy Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, 14194, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Yang X, Fox A, DeCarlo C, Pineda N, Powell RL. The Secretory IgA Response in Human Milk Against the SARS-CoV-2 Spike Is Highly Durable and Neutralizing for At Least 1 Year of Lactation Postinfection. Breastfeed Med 2023; 18:602-611. [PMID: 37615565 PMCID: PMC10460685 DOI: 10.1089/bfm.2023.0117] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Background: Although in the early pandemic period COVID-19 pathology among young children and infants was typically less severe compared with that observed among adults, this has not remained entirely consistent as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants have emerged. There is an enormous body of evidence demonstrating the benefits of human milk antibodies (Abs) in protecting infants against a wide range of enteric and respiratory infections. It is highly plausible that the same holds true for protection against SARS-CoV-2 as this virus infects cells of the gastrointestinal and respiratory mucosae. Understanding the durability of a human milk Ab response over time after infection is critical. Objective: Previously, we examined the Abs present in milk of those recently infected with SARS-CoV-2 and concluded that the response was secretory immunoglobulin A (sIgA) dominant and that these titers were highly correlated with neutralization potency. The present study aimed to monitor the durability of the SARS-CoV-2 IgA and secretory Ab (sAb) response in milk from COVID-19-recovered lactating individuals over 12 months in the absence of vaccination or reinfection. Results: This analysis revealed a robust and durable spike-specific milk sIgA response, and at 9-12 months after infection, 88% of the samples exhibited titers above the positive cutoff for IgA and 94% were above the cutoff for sAb. Fifty percent of participants exhibited less than twofold reduction of spike-specific IgA through 12 months. A strong, significant positive correlation between IgA and sAb against spike persisted throughout the study period. Nucleocapsid-specific Abs were also assessed, which revealed significant background or cross-reactivity of milk IgA against this immunogen, as well as limited/inconsistent durability compared with Spike titers. Conclusion: These data suggest that lactating individuals are likely to continue producing spike-specific Abs in their milk for 1 year or more, which may provide critical passive immunity to infants against SARS-CoV-2 throughout the lactation period.
Collapse
Affiliation(s)
- Xiaoqi Yang
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alisa Fox
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Claire DeCarlo
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nicole Pineda
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rebecca L.R. Powell
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
22
|
Ware J, McElhinney K, Latham T, Lane A, Dienger-Stambaugh K, Hildeman D, Spearman P, Ware RE. Sustained and Boosted Antibody Responses in Breast Milk After Maternal SARS-CoV-2 Vaccination. Breastfeed Med 2023; 18:612-620. [PMID: 37615566 DOI: 10.1089/bfm.2023.0106] [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] [Indexed: 08/25/2023]
Abstract
Background: Pregnant and lactating women were not included in the initial large vaccine clinical trials for SARS-CoV-2 (COVID) infection. Delineating the antibody titers in serum and breast milk of lactating women is important to determine the safety and benefits of vaccination in this special population. Objective: To investigate COVID vaccinations in breastfeeding dyads and effects on lactation, the Antibody Detection of Vaccine-Induced Secretory Effects trial (ADVISE) prospectively evaluated anti-COVID antibodies in serum and breast milk after initial paired and booster vaccines. Methods: This is a prospective longitudinal surveillance cohort study of lactating women. Eligibility criteria included ≥18 years of age, currently lactating, and at enrollment either received COVID vaccination within the past 60 days or planning vaccination within 60 days. Results: Among 63 lactating mothers, COVID vaccination led to breast milk secretory IgA (sIgA) and IgG antibodies with consistent viral neutralizing activity. Milk sIgA titers increased further after second vaccination and were prolonged after a third booster dose, including women with extended breastfeeding beyond 12 months. Milk IgG antibody titers were higher and more sustained than sIgA. Antibody titers were not associated with individual dyad characteristics or vaccine manufacturer. Vaccine-induced antibodies from milk were not detected in infant circulation. Conclusions and Relevance: Maternal COVID vaccination during lactation is well tolerated and generates sustained and boosted antibody responses in breast milk. COVID-specific sIgA and IgG antibodies with neutralizing activity are found in breast milk, including boosted mothers who continue breastfeeding beyond 12 months. These data support universal COVID vaccinations for all lactating mothers, including booster immunizations during extended breastfeeding (NCT04895475).
Collapse
Affiliation(s)
- Julie Ware
- Division of General and Community Pediatrics, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kathryn McElhinney
- Division of Hematology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Teresa Latham
- Division of Hematology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Adam Lane
- Division of Hematology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Krista Dienger-Stambaugh
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - David Hildeman
- Division of Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Paul Spearman
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Russell E Ware
- Division of Hematology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
23
|
Bäuerl C, Zulaica J, Rusu L, Moreno AR, Pérez-Cano FJ, Lerin C, Mena-Tudela D, Aguilar-Camprubí L, Parra-Llorca A, Martínez-Costa C, Geller R, Collado MC. Assessment of SARS-CoV-2 neutralizing antibody titers in breastmilk from convalescent and vaccinated mothers. iScience 2023; 26:106802. [PMID: 37197591 PMCID: PMC10158041 DOI: 10.1016/j.isci.2023.106802] [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: 02/16/2023] [Revised: 03/29/2023] [Accepted: 04/28/2023] [Indexed: 05/19/2023] Open
Abstract
Breastmilk contains antibodies that could protect breastfed infants from infections. In this work, we examined if antibodies in breastmilk could neutralize SARS-CoV-2 in 84 breastmilk samples from women that were either vaccinated (Comirnaty, mRNA-1273, or ChAdOx1), infected with SARS-CoV-2, or both infected and vaccinated. The neutralization capacity of these sera was tested using pseudotyped vesicular stomatitis virus carrying either the Wuhan-Hu-1, Delta, or BA.1 Omicron spike proteins. We found that natural infection resulted in higher neutralizing titers and that neutralization correlated positively with levels of immunoglobulin A in breastmilk. In addition, significant differences in the capacity to produce neutralizing antibodies were observed between both mRNA-based vaccines and the adenovirus-vectored ChAdOx1 COVID-19 vaccine. Overall, our results indicate that breastmilk from naturally infected women or those vaccinated with mRNA-based vaccines contains SARS-CoV-2 neutralizing antibodies that could potentially provide protection to breastfed infants from infection.
Collapse
Affiliation(s)
- Christine Bäuerl
- Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), 46980 Paterna, Valencia, Spain
| | - Joao Zulaica
- Institute for Integrative Systems Biology (ISysBio), University of Valencia-CSIC, 46980 Paterna, Valencia, Spain
| | - Luciana Rusu
- Institute for Integrative Systems Biology (ISysBio), University of Valencia-CSIC, 46980 Paterna, Valencia, Spain
| | - Alicia Rodríguez Moreno
- Institute for Integrative Systems Biology (ISysBio), University of Valencia-CSIC, 46980 Paterna, Valencia, Spain
| | - Francisco J. Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science and Institute of Research in Nutrition and Food Safety (INSA), University of Barcelona (UB), 08028 Barcelona, Spain
| | - Carles Lerin
- Endocrinology department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Desirée Mena-Tudela
- Department of Nursing, Nursing Research Group, Universitat Jaume I, Castellón, Spain
| | | | - Anna Parra-Llorca
- Health Research Institute La Fe, Neonatal Research Group, Spain and University and Polytechnic Hospital La Fe, Division of Neonatology, 46026 Valencia, Spain
| | - Cecilia Martínez-Costa
- Department of Pediatrics, Hospital Clínico Universitario, University of Valencia, Spain. Nutrition Research Group of INCLIVA, 46010 Valencia, Spain
| | - Ron Geller
- Institute for Integrative Systems Biology (ISysBio), University of Valencia-CSIC, 46980 Paterna, Valencia, Spain
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), 46980 Paterna, Valencia, Spain
| | - MilkCORONA study team
- Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), 46980 Paterna, Valencia, Spain
- Institute for Integrative Systems Biology (ISysBio), University of Valencia-CSIC, 46980 Paterna, Valencia, Spain
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science and Institute of Research in Nutrition and Food Safety (INSA), University of Barcelona (UB), 08028 Barcelona, Spain
- Endocrinology department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Nursing, Nursing Research Group, Universitat Jaume I, Castellón, Spain
- LactApp Women Health, Barcelona, Spain
- Health Research Institute La Fe, Neonatal Research Group, Spain and University and Polytechnic Hospital La Fe, Division of Neonatology, 46026 Valencia, Spain
- Department of Pediatrics, Hospital Clínico Universitario, University of Valencia, Spain. Nutrition Research Group of INCLIVA, 46010 Valencia, Spain
| |
Collapse
|
24
|
Briana DD, Malamitsi-Puchner A. Breastfeeding provides a protective hug and the benefits have outweighed the risks during the COVID-19 pandemic. Acta Paediatr 2023; 112:1177-1181. [PMID: 36945791 DOI: 10.1111/apa.16769] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/18/2023] [Accepted: 03/20/2023] [Indexed: 03/23/2023]
Abstract
Mothers have been very hesitant about breastfeeding when they have COVID-19 infection or vaccinations. Maternal milk protects neonates through its high biological value, immune factors and anti-infectious molecules and this review shows that the virus that causes COVID-19 is not transmitted through breast milk. COVID-19 vaccines induce anti-spike antibodies with neutralising capacity, and phagocytosis, and no vaccine particles or messenger ribonucleic acid have been detected in breast milk. Most drugs used for maternal COVID-19 infections are safe for breastfed infants. CONCLUSION: The clear benefits of breastfeeding by far outweigh the very low risk of infant infections from COVID-19.
Collapse
Affiliation(s)
- Despina D Briana
- Neonatal Intensive Care Unit, 3rd Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| | - Ariadne Malamitsi-Puchner
- Neonatal Intensive Care Unit, 3rd Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
25
|
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: 0] [Impact Index Per Article: 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
|
26
|
Stafford LS, Valcarce V, Henry M, Neu J, Parker L, Mueller M, Vicuna V, Gowen T, Cato E, Kosik I, Yewdell JW, Atkinson M, Cacho N, Li N, Larkin J. Detection of SARS-CoV-2 IgA and IgG in human milk and breastfeeding infant stool 6 months after maternal COVID-19 vaccination. J Perinatol 2023; 43:775-781. [PMID: 36631564 DOI: 10.1038/s41372-022-01581-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/28/2022] [Accepted: 12/05/2022] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Assess presence, durability, and neutralization capacity of SARS-CoV-2-specific antibodies in breastfeeding infants' stool, mother's plasma and milk following maternal vaccination. DESIGN Thirty-seven mothers and 25 infants were enrolled between December 2020 and November 2021 for this prospective observational study. All mothers were vaccinated during lactation except three, which were vaccinated during pregnancy. Milk, maternal plasma, and infants' stool was collected pre-vaccination and at periods up to 6 months following COVID-19 vaccine series initiation/completion. SARS-CoV-2 antibody levels and their neutralization capacities were assessed. RESULTS SARS-CoV-2-specific IgA and IgG levels were higher in infant stool post-maternal vaccination amongst milk-fed compared to controls. Maternal SARS-CoV-2-specific IgA and IgG concentrations decreased over 6 months post-vaccination but remained higher than pre-vaccination levels. We observed improved neutralization capacity in milk and plasma after COVID-19 vaccination. CONCLUSIONS The presence of SARS-CoV-2-specific antibodies in infant stool following maternal vaccination offers further evidence of the lasting transfer of these antibodies through breastfeeding.
Collapse
Affiliation(s)
| | - Vivian Valcarce
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Matthew Henry
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Josef Neu
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Leslie Parker
- College of Nursing, University of Florida, Gainesville, FL, USA
| | - Martina Mueller
- College of Nursing, Medical University of South Carolina, Charleston, SC, USA
| | - Valeria Vicuna
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, USA
| | - Taylor Gowen
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Emilee Cato
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - Ivan Kosik
- Laboratory of Viral Diseases, National Institutes of Health/National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Jonathan Wilson Yewdell
- Laboratory of Viral Diseases, National Institutes of Health/National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Mark Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Nicole Cacho
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Nan Li
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Joseph Larkin
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
27
|
Yang X, Fox A, DeCarlo C, Pineda N, Powell RL. The secretory IgA (sIgA) response in human milk against the SARS-CoV-2 Spike is highly durable and neutralizing for at least 1 year of lactation post-infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.19.23290192. [PMID: 37293109 PMCID: PMC10246141 DOI: 10.1101/2023.05.19.23290192] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Although in the early pandemic period, COVID-19 pathology among young children and infants was typically less severe compared to that observed among adults, this has not remained entirely consistent as SARS-CoV-2 variants have emerged. There is an enormous body of evidence demonstrating the benefits of human milk antibodies (Abs) in protecting infants against a wide range of enteric and respiratory infections. It is highly plausible that the same holds true for protection against SARS-CoV-2, as this virus infects cells of the gastrointestinal and respiratory mucosae. Understanding the durability of a human milk Ab response over time after infection is critical. Previously, we examined the Abs present in milk of those recently infected with SARS-CoV-2, and concluded that the response was secretory IgA (sIgA)-dominant and that these titers were highly correlated with neutralization potency. The present study aimed to monitor the durability of the SARS-CoV-2 IgA and secretory Ab (sAb) response in milk from COVID-19-recovered lactating individuals over 12 months, in the absence of vaccination or re-infection. This analysis revealed a robust and durable Spike-specific milk sIgA response, that at 9-12 months after infection, 88% of the samples exhibited titers above the positive cutoff for IgA and 94% were above cutoff for sAb. Fifty percent of participants exhibited less than a 2-fold reduction of Spike-specific IgA through 12 months. A strong significant positive correlation between IgA and sAb against Spike persisted throughout the study period. Nucleocapsid-specific Abs were also assessed, which revealed significant background or cross reactivity of milk IgA against this immunogen, as well as limited/inconsistent durability compared to Spike titers. These data suggests that lactating individuals are likely to continue producing Spike-specific Abs in their milk for 1 year or more, which may provide critical passive immunity to infants against SARS-CoV-2 throughout the lactation period.
Collapse
Affiliation(s)
- Xiaoqi Yang
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York, USA
| | - Alisa Fox
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York, USA
| | - Claire DeCarlo
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York, USA
| | - Nicole Pineda
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York, USA
| | - Rebecca L.R. Powell
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York, USA
| |
Collapse
|
28
|
Maniu I, Maniu GC, Antonescu E, Duica L, Grigore N, Totan M. SARS-CoV-2 Antibody Responses in Pediatric Patients: A Bibliometric Analysis. Biomedicines 2023; 11:biomedicines11051455. [PMID: 37239126 DOI: 10.3390/biomedicines11051455] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
The characteristics, dynamics and mechanisms/determinants of the immune response to SARS-CoV-2 infection are not fully understood. We performed a bibliometric review of studies that have assessed SARS-CoV-2 antibody responses in the pediatric population using Web of Science online databases, VOSviewer and Bibliometrix tools. The analysis was conducted on 84 publications, from 310 institutions located in 29 countries and published in 57 journals. The results showed the collaboration of scientists and organizations, international research interactions and summarized the findings on (i) the measured titers of antibodies (total antibody and/or individual antibody classes IgG, IgM, IgA) against different antigens (C-terminal region of N (N CT), full-length N protein (N FL), RBD, RBD Alpha, RBD Beta, RBD Gamma, RBD Delta, spike (S), S1, S2) in the case of different clinical forms of the disease; and (ii) the correlations between SARS-CoV-2 antibodies and cytokines, chemokines, neutrophils, C-reactive protein, ferritin, and the erythrocyte sedimentation rate. The presented study offers insights regarding research directions to be explored in the studied field and may provide a starting point for future research.
Collapse
Affiliation(s)
- Ionela Maniu
- Mathematics and Informatics Department, Research Center in Informatics and Information Technology, Faculty of Sciences, "Lucian Blaga" University, 5-7 Ion Ratiu Str., 550025 Sibiu, Romania
- Pediatric Research Team, Clinical Pediatric Hospital, 2-4 Pompeiu Onofreiu Str., 550166 Sibiu, Romania
| | - George Constantin Maniu
- Mathematics and Informatics Department, Research Center in Informatics and Information Technology, Faculty of Sciences, "Lucian Blaga" University, 5-7 Ion Ratiu Str., 550025 Sibiu, Romania
| | - Elisabeta Antonescu
- Faculty of Medicine, Lucian Blaga University of Sibiu, 2A Lucian Blaga Str., 550169 Sibiu, Romania
- County Clinical Emergency Hospital, 2-4 Corneliu Coposu Str., 550245 Sibiu, Romania
| | - Lavinia Duica
- Faculty of Medicine, Lucian Blaga University of Sibiu, 2A Lucian Blaga Str., 550169 Sibiu, Romania
- County Clinical Emergency Hospital, 2-4 Corneliu Coposu Str., 550245 Sibiu, Romania
| | - Nicolae Grigore
- Faculty of Medicine, Lucian Blaga University of Sibiu, 2A Lucian Blaga Str., 550169 Sibiu, Romania
- County Clinical Emergency Hospital, 2-4 Corneliu Coposu Str., 550245 Sibiu, Romania
| | - Maria Totan
- Faculty of Medicine, Lucian Blaga University of Sibiu, 2A Lucian Blaga Str., 550169 Sibiu, Romania
- Clinical Laboratory, Clinical Pediatric Hospital, 2-4 Pompeiu Onofreiu Str., 550166 Sibiu, Romania
| |
Collapse
|
29
|
Dimitroglou M, Sokou R, Iacovidou N, Pouliakis A, Kafalidis G, Boutsikou T, Iliodromiti Z. Anti-SARS-CoV-2 Immunoglobulins in Human Milk after Coronavirus Disease or Vaccination-Time Frame and Duration of Detection in Human Milk and Factors That Affect Their Titers: A Systematic Review. Nutrients 2023; 15:nu15081905. [PMID: 37111124 PMCID: PMC10141636 DOI: 10.3390/nu15081905] [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: 03/06/2023] [Revised: 03/31/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Human milk (HM) of mothers infected with or vaccinated against SARS-CoV-2 contains specific immunoglobulins, which may protect their offspring against infection or severe disease. The time frame and duration after infection or vaccination, during which these immunoglobulins are detected in HM, as well as the major factors that influence their levels, have not been fully elucidated. This systematic review aimed to collect the existing literature and describe the immune response, specifically regarding the immunoglobulins in HM after COVID-19 disease or vaccination in non-immune women. We conducted a systematic search of PubMed and Scopus databases to identify studies published up until 19 March 2023. In total, 975 articles were screened, and out of which 75 were identified as being relevant and were finally included in this review. Infection by SARS-CoV-2 virus primarily induces an IgA immune response in HM, while vaccination predominantly elevates IgG levels. These immunoglobulins give HM a neutralizing capacity against SARS-CoV-2, highlighting the importance of breastfeeding during the pandemic. The mode of immune acquisition (infection or vaccination) and immunoglobulin levels in maternal serum are factors that seem to influence immunoglobulin levels in HM. Further studies are required to determine the impact of other factors, such as infection severity, lactation period, parity, maternal age and BMI on immunoglobulin level in HM.
Collapse
Affiliation(s)
- Margarita Dimitroglou
- Neonatal Department, Medical School, National and Kapodistrian University of Athens, Aretaieio Hospital, 11528 Athens, Greece
| | - Rozeta Sokou
- Neonatal Department, Medical School, National and Kapodistrian University of Athens, Aretaieio Hospital, 11528 Athens, Greece
| | - Nicoletta Iacovidou
- Neonatal Department, Medical School, National and Kapodistrian University of Athens, Aretaieio Hospital, 11528 Athens, Greece
| | - Abraham Pouliakis
- Neonatal Department, Medical School, National and Kapodistrian University of Athens, Aretaieio Hospital, 11528 Athens, Greece
| | - Georgios Kafalidis
- Neonatal Department, Medical School, National and Kapodistrian University of Athens, Aretaieio Hospital, 11528 Athens, Greece
| | - Theodora Boutsikou
- Neonatal Department, Medical School, National and Kapodistrian University of Athens, Aretaieio Hospital, 11528 Athens, Greece
| | - Zoi Iliodromiti
- Neonatal Department, Medical School, National and Kapodistrian University of Athens, Aretaieio Hospital, 11528 Athens, Greece
| |
Collapse
|
30
|
Ramaswamy VV, Abiramalatha T, Pullattayil S AK, Trevisanuto D. Multisystem inflammatory disease in neonates (MIS-N) due to maternal COVID-19. Semin Fetal Neonatal Med 2023; 28:101431. [PMID: 37061425 DOI: 10.1016/j.siny.2023.101431] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2023]
Abstract
Multisystem inflammatory disease in neonates (MIS-N) is a disease of immune dysregulation presenting in the newborn period. Thouvgh its etiopathogenesis is proposed to be similar to multisystem inflammatory disease in Children (MIS-C), the exact pathophysiology is largely unknown as of present. The definition of MIS-N is contentious. The evidence for its incidence, the clinical features, profile of raised inflammatory markers, treatment strategies and outcomes stem from case reports, case series and cohort studies with small sample sizes. Though the incidence of MIS-N in severe acute respiratory syndrome caused by the coronavirus CoVID-2 (SARS-CoV-2) infected asymptomatic neonates is low, its incidence in symptomatic neonates is relatively higher. Further, amongst the neonates who are treated as MIS-N, the mortality rate is high. The review also evaluates the various other unresolved aspects of MIS-N from limited published literature and identifies knowledge gaps which could be areas of future research.
Collapse
Affiliation(s)
| | | | | | - Daniele Trevisanuto
- Department of Woman's and Child's Health, University Hospital of Padua, Padua, Italy.
| |
Collapse
|
31
|
Agostinis C, Toffoli M, Balduit A, Mangogna A, Yasmin H, Ragazzon C, Pegoraro S, Campisciano G, Stabile G, Zito G, Kishore U, Comar M, Scrimin F, Bulla R, Ricci G. Anti-Spike Antibodies Present in the Milk of SARS-CoV-2 Vaccinated Mothers Are Complement-Activating. Int J Mol Sci 2023; 24:4395. [PMID: 36901824 PMCID: PMC10002545 DOI: 10.3390/ijms24054395] [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: 12/29/2022] [Revised: 02/02/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Although only 0.8-1% of SARS-CoV-2 infections are in the 0-9 age-group, pneumonia is still the leading cause of infant mortality globally. Antibodies specifically directed against SARS-CoV-2 spike protein (S) are produced during severe COVID-19 manifestations. Following vaccination, specific antibodies are also detected in the milk of breastfeeding mothers. Since antibody binding to viral antigens can trigger activation of the complement classical - pathway, we investigated antibody-dependent complement activation by anti-S immunoglobulins (Igs) present in breast milk following SARS-CoV-2 vaccination. This was in view of the fact that complement could play a fundamentally protective role against SARS-CoV-2 infection in newborns. Thus, 22 vaccinated, lactating healthcare and school workers were enrolled, and a sample of serum and milk was collected from each woman. We first tested for the presence of anti-S IgG and IgA in serum and milk of breastfeeding women by ELISA. We then measured the concentration of the first subcomponents of the three complement pathways (i.e., C1q, MBL, and C3) and the ability of anti-S Igs detected in milk to activate the complement in vitro. The current study demonstrated that vaccinated mothers have anti-S IgG in serum as well as in breast milk, which is capable of activating complement and may confer a protective benefit to breastfed newborns.
Collapse
Affiliation(s)
- Chiara Agostinis
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy
| | - Miriam Toffoli
- Department of Medical, Surgical and Health Science, University of Trieste, 34129 Trieste, Italy
| | - Andrea Balduit
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy
| | - Alessandro Mangogna
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy
| | - Hadida Yasmin
- Immunology and Cell Biology Laboratory, Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar 736101, West Bengal, India
| | - Chiara Ragazzon
- Department of Medical, Surgical and Health Science, University of Trieste, 34129 Trieste, Italy
| | - Silvia Pegoraro
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy
| | | | - Guglielmo Stabile
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy
| | - Gabriella Zito
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy
| | - Uday Kishore
- Department of Veterinary Medicine, United Arab Emirates University (UAEU), Al Ain P.O. Box 15551, United Arab Emirates
| | - Manola Comar
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy
- Department of Medical, Surgical and Health Science, University of Trieste, 34129 Trieste, Italy
| | - Federica Scrimin
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Giuseppe Ricci
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy
- Department of Medical, Surgical and Health Science, University of Trieste, 34129 Trieste, Italy
| |
Collapse
|
32
|
Guan B, Chai Y, Amantai X, Liu X, Chen X, Cao X, Yue X, Liu B. Glycoproteomics analysis reveals differential site-specific N-glycosylation of donkey milk fat globule membrane protein during lactation. Food Chem 2023; 402:134266. [DOI: 10.1016/j.foodchem.2022.134266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 11/29/2022]
|
33
|
Evaluation of Anti-SARS-CoV-2 IgA Response in Tears of Vaccinated COVID-19 Subjects. Viruses 2023; 15:v15020399. [PMID: 36851613 PMCID: PMC9965053 DOI: 10.3390/v15020399] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/17/2023] [Accepted: 01/28/2023] [Indexed: 02/01/2023] Open
Abstract
Secretory IgA (sIgA), which may play an important role in the early defense against SARS-CoV-2 infection, were detected in the eye of COVID-19 patients. However, an evaluation of the sIgA response in the tears of vaccinated or non-vaccinated COVID-19 subjects is still lacking. Aimed at characterizing sIgA mucosal immunity in the eye, this study analyzed tear samples from 77 COVID-19 patients, including 63 vaccinated and 14 non-vaccinated subjects. The groups showed similar epidemiological features, but as expected, differences were observed in the percentage of asymptomatic/pauci-symptomatic subjects in the vaccinated vs. non-vaccinated cohort (46% and 29% of the total, respectively). Consistent with this, ocular sIgA values, evaluated by a specific quantitative ELISA assay, were remarkably different in vaccinated vs. non-vaccinated group for both frequency (69.8% vs. 57.1%, respectively) and titer (1372.3 U/mL vs. 143.7 U/mL, respectively; p = 0.01), which was significantly differently elevated depending on the type of administered vaccine. The data show for the first time significant differences of available vaccines to elicit sIgA response in the eye and suggest that quantitative tear-based sIgA tests may potentially serve as a rapid and easily accessible biomarker for the assessment of the development of a protective mucosal immunity toward SARS-CoV-2.
Collapse
|
34
|
Gribble K, Cashin J, Marinelli K, Vu DH, Mathisen R. First do no harm overlooked: Analysis of COVID-19 clinical guidance for maternal and newborn care from 101 countries shows breastfeeding widely undermined. Front Nutr 2023; 9:1049610. [PMID: 36741988 PMCID: PMC9889271 DOI: 10.3389/fnut.2022.1049610] [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: 09/20/2022] [Accepted: 12/14/2022] [Indexed: 01/19/2023] Open
Abstract
Background In March 2020, the World Health Organization (WHO) published clinical guidance for the care of newborns of mothers with COVID-19. Weighing the available evidence on SARS-CoV-2 infection against the well-established harms of maternal-infant separation, the WHO recommended maternal-infant proximity and breastfeeding even in the presence of maternal infection. Since then, the WHO's approach has been validated by further research. However, early in the pandemic there was poor global alignment with the WHO recommendations. Methods We assessed guidance documents collected in November and December 2020 from 101 countries and two regional agencies on the care of newborns of mothers with COVID-19 for alignment with the WHO recommendations. Recommendations considered were: (1) skin-to-skin contact; (2) early initiation of breastfeeding; (3) rooming-in; (4) direct breastfeeding; (5) provision of expressed breastmilk; (6) provision of donor human milk; (7) wet nursing; (8) provision of breastmilk substitutes; (9) relactation; (10) psychological support for separated mothers; and (11) psychological support for separated infants. Results In less than one-quarter of country guidance were the three key breastfeeding facilitation practices of skin-to-skin contact, rooming-in, and direct breastfeeding recommended. Donor human milk was recommended in under one-quarter of guidance. Psychological support for mothers separated from their infants was recommended in 38%. Few countries recommended relactation, wet nursing, or psychological support for infants separated from mothers. In three-quarters of country guidance, expressed breastmilk for infants unable to directly breastfeed was recommended. The WHO and the United Kingdom's Royal College of Obstetricians and Gynecologists were each cited by half of country guidance documents with the United States Centers for Disease Control and Prevention directly or indirectly cited by 40%. Conclusion Despite the WHO recommendations, many COVID-19 maternal and newborn care guidelines failed to recommend skin-to-skin contact, rooming-in, and breastfeeding as the standard of care. Irregular guidance updates and the discordant, but influential, guidance from the United States Centers for Disease Control may have been contributory. It appeared that once recommendations were made for separation or against breastfeeding they were difficult to reverse. In the absence of quality evidence on necessity, recommendations against breastfeeding should not be made in disease epidemics.
Collapse
Affiliation(s)
- Karleen Gribble
- School of Nursing and Midwifery, Western Sydney University, Parramatta, NSW, Australia
| | - Jennifer Cashin
- Alive & Thrive Southeast Asia, FHI 360, Washington, DC, United States
| | - Kathleen Marinelli
- Department of Pediatrics, University of Connecticut School of Medicine, Hartford, CT, United States
| | | | | |
Collapse
|
35
|
Cambou MC, Liu CM, Mok T, Fajardo-Martinez V, Paiola SG, Ibarrondo FJ, Kerin T, Fuller T, Tobin NH, Garcia G, Bhattacharya D, Aldrovandi GM, Arumugaswami V, Foo SS, Jung JU, Vasconcelos Z, Brasil P, Brendolin M, Yang O, Rao R, Nielsen-Saines K. Longitudinal Evaluation of Antibody Persistence in Mother-Infant Dyads After Severe Acute Respiratory Syndrome Coronavirus 2 Infection in Pregnancy. J Infect Dis 2023; 227:236-245. [PMID: 36082433 PMCID: PMC9494415 DOI: 10.1093/infdis/jiac366] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/01/2022] [Accepted: 09/06/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND There are limited data on how coronavirus disease 2019 (COVID-19) severity, timing of infection, and subsequent vaccination impact transplacental transfer and persistence of maternal and infant antibodies. METHODS In a longitudinal cohort of pregnant women with polymerase chain reaction-confirmed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, maternal/infant sera were collected at enrollment, delivery/birth, and 6 months. Anti-SARS-CoV-2 spike immunoglobulin (Ig)G, IgM, and IgA were measured by enzyme-linked immunosorbent assay. RESULTS Two-hundred fifty-six pregnant women and 135 infants were enrolled; 148 maternal and 122 neonatal specimens were collected at delivery/birth; 45 maternal and 48 infant specimens were collected at 6 months. Sixty-eight percent of women produced all anti-SARS-CoV-2 isotypes at delivery (IgG, IgM, IgA); 96% had at least 1 isotype. Symptomatic disease and vaccination before delivery were associated with higher maternal IgG at labor and delivery. Detectable IgG in infants dropped from 78% at birth to 52% at 6 months. In the multivariate analysis evaluating factors associated with detectable IgG in infants at delivery, significant predictors were 3rd trimester infection (odds ratio [OR] = 4.0), mild/moderate disease (OR = 4.8), severe/critical disease (OR = 6.3), and maternal vaccination before delivery (OR = 18.8). No factors were significant in the multivariate analysis at 6 months postpartum. CONCLUSIONS Vaccination in pregnancy post-COVID-19 recovery is a strategy for boosting antibodies in mother-infant dyads.
Collapse
Affiliation(s)
- Mary C Cambou
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, U.S
| | - Christine M Liu
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, U.S
| | - Thalia Mok
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, U.S
| | - Viviana Fajardo-Martinez
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, U.S
| | - Sophia G Paiola
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, U.S
| | - Francisco J Ibarrondo
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, U.S
| | - Tara Kerin
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, U.S
| | - Trevon Fuller
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, U.S
- Fundação Oswaldo Cruz, Rio de Janeiro, RJ, 21040360, Brazil
| | - Nicole H Tobin
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, U.S
| | - Gustavo Garcia
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, U.S
| | - Debika Bhattacharya
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, U.S
| | - Grace M Aldrovandi
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, U.S
| | | | - Suan Sin Foo
- Department of Cancer Biology, Infection Biology Program, and Global Center for Pathogen and Human Health Research, Cleveland Clinic, Cleveland, OH, 44195, U.S
| | - Jae U Jung
- Department of Cancer Biology, Infection Biology Program, and Global Center for Pathogen and Human Health Research, Cleveland Clinic, Cleveland, OH, 44195, U.S
| | | | | | - Michelle Brendolin
- Maternidade do Hospital Estadual Adão Pereira Nunes, Caxias, RJ, 25221-970, Brazil
| | - Otto Yang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, U.S
| | - Rashmi Rao
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, U.S
| | - Karin Nielsen-Saines
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, U.S
| |
Collapse
|
36
|
Puerta A, Garcia-Lopez D, Tejedor-Matellanes P, Gomez-Ruiz L, de la Cruz-Rodriguez R, de Frutos M. Capillary gel electrophoresis of very high molecular weight glycoproteins. Commercial and tailor-made gels for analysis of human monomeric and secretory immunoglobulin A. J Chromatogr A 2023; 1688:463689. [PMID: 36528901 DOI: 10.1016/j.chroma.2022.463689] [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: 07/14/2022] [Revised: 10/18/2022] [Accepted: 11/27/2022] [Indexed: 12/07/2022]
Abstract
Capillary gel electrophoresis (CGE) has been widely used for analysis of proteins according to their size. However, to our knowledge, this technique has not been optimized to immunoglobulin A (IgA) analysis, a protein of current and emerging high interest in several fields. IgA is the first barrier of human body against pathogens. This protein in human milk and colostrum is essential for immune protection of newborns and treatment of milk for storage in Human Milk Banks may alter IgA. The emerging use of IgA as therapeutic treatment also encourages the development of analysis methods for this class of immunoglobulins. IgA is far more heterogeneously glycosylated and complex than the well-studied IgG molecules. IgA in serum is mainly monomeric (mIgA) with about 160 kDa, while in secretions such as saliva, milk, colostrum, etc, secretory immunoglobulin A (sIgA) is the predominant form. This is a dimer where both monomers are linked by the J-chain and the secretory component accounting all together for a MW higher than 400 kDa including the glycans. This size is far from the 225 kDa MW for which commercial CGE kits are intended. The general rules governing CGE behavior of analytes cannot be directly applied to every protein. Addressing studies directed specifically to target proteins is specially needed for the large size and highly complex target analytes of this study. In this work the effect of several factors on CGE analysis of human serum and colostrum IgA is studied. The feasibility of performing analysis of both IgA classes using a commercial CGE kit is shown. In addition, this work introduces another novelty by preparing tailor-made reproducible gel buffers and to characterize them in terms of dynamic viscosity, conductivity, and electroosmotic flow mobility in bare fused silica capillaries. The possibility of analyzing mIgA and sIgA in less than 10 min using these tailor-made gels is demonstrated. Inter-day variation (RSD) for the main peak of sIgA is 0.25% for migration time (tm) and 0.27% for percentage corrected peak area (Acorr).
Collapse
Affiliation(s)
- Angel Puerta
- Institute of Organic Chemistry (IQOG-CSIC), Juan de la Cierva 3, E-28006 Madrid, Spain.
| | - Daniel Garcia-Lopez
- Institute of Organic Chemistry (IQOG-CSIC), Juan de la Cierva 3, E-28006 Madrid, Spain
| | | | - Laura Gomez-Ruiz
- Institute of Organic Chemistry (IQOG-CSIC), Juan de la Cierva 3, E-28006 Madrid, Spain
| | | | - Mercedes de Frutos
- Institute of Organic Chemistry (IQOG-CSIC), Juan de la Cierva 3, E-28006 Madrid, Spain
| |
Collapse
|
37
|
Graciliano NG, Tenório MCS, Fragoso MBT, Moura FA, Botelho RM, Tanabe ELL, Borbely KSC, Borbely AU, Oliveira ACM, Goulart MOF. The impact on colostrum oxidative stress, cytokines, and immune cells composition after SARS-CoV-2 infection during pregnancy. Front Immunol 2022; 13:1031248. [PMID: 36591280 PMCID: PMC9798093 DOI: 10.3389/fimmu.2022.1031248] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Background Limited data are available regarding the differences between immunological, biochemical, and cellular contents of human colostrum following maternal infection during pregnancy with coronavirus 2 disease (COVID-19). Objective To investigate whether maternal COVID-19 infection may affect immunological, biochemical, and cellular contents of human colostrum. Methods Using a case-control study design, we collected colostrum from 14 lactating women with a previous diagnosis of COVID-19 during pregnancy and 12 without a clear diagnosis during September 2020 to May 2021. Colostrum samples were analysed for some enzymes and non-enzymatic oxidative stress markers (SOD, CAT, GPx, MDA, GSH, GSSG, H2O2, MPO) and for IL-1β, IL-6, tumour necrosis factor (TNF)-α, protein induced by interferon gamma (IP)-10, IL-8, IFN-λ1, IL12p70, IFN-α2, IFN-λ2/3, granulocyte macrophage colony stimulating factor (GM-CSF), IFN-β, IL-10 and IFN-γ, along with IgA and IgG for the SARS-CoV-2 S protein. We perform immunophenotyping to assess the frequency of different cell types in the colostrum. Results Colostrum from the COVID-19 symptomatic group in pregnancy contained reduced levels of H2O2, IFN-α2, and GM-CSF. This group had higher levels of GSH, and both NK cell subtypes CD3-CD56brightCD16-CD27+IFN-γ+ and CD3-CD56dimCD16+CD27- were also increased. Conclusion The present results reinforce the protective role of colostrum even in the case of mild SARS-Cov-2 infection, in addition to demonstrating how adaptive the composition of colostrum is after infections. It also supports the recommendation to encourage lactating women to continue breastfeeding after COVID-19 illness.
Collapse
Affiliation(s)
- Nayara Gomes Graciliano
- Institute of Biological and Health Sciences, Federal University of Alagoas, Maceio, Alagoas, Brazil
| | | | | | | | - Rayane Martins Botelho
- Institute of Biological and Health Sciences, Federal University of Alagoas, Maceio, Alagoas, Brazil
| | - Eloiza Lopes Lira Tanabe
- Institute of Biological and Health Sciences, Federal University of Alagoas, Maceio, Alagoas, Brazil
| | | | - Alexandre Urban Borbely
- Institute of Biological and Health Sciences, Federal University of Alagoas, Maceio, Alagoas, Brazil
| | - Alane Cabral Menezes Oliveira
- Institute of Biological and Health Sciences, Federal University of Alagoas, Maceio, Alagoas, Brazil
- College of Nutrition, Federal University of Alagoas, Maceio, Alagoas, Brazil
| | - Marília Oliveira Fonseca Goulart
- Institute of Biological and Health Sciences, Federal University of Alagoas, Maceio, Alagoas, Brazil
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceio, Alagoas, Brazil
- National Institute of Science and Technology in Bioanalytics (INCT-Bio), Campinas, Sao Paulo, Brazil
| |
Collapse
|
38
|
Golan Y, Ilala M, Gay C, Hunagund S, Lin CY, Cassidy AG, Jigmeddagva U, Li L, Ozarslan N, Asiodu IV, Ahituv N, Flaherman VJ, Gaw SL, Prahl M. Milk antibody response after 3rd dose of COVID-19 mRNA vaccine and SARS-CoV-2 breakthrough infection and implications for infant protection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.12.12.22283367. [PMID: 36561188 PMCID: PMC9774223 DOI: 10.1101/2022.12.12.22283367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Anti-SARS-CoV-2 antibodies have been found in human-milk after COVID-19 infection and vaccination. However, little is known about their persistence in milk after booster vaccination and breakthrough infection. In this study, human-milk, saliva and blood samples were collected from 33 lactating individuals before and after mRNA-based vaccination and COVID-19 breakthrough infections. Antibody levels were measured using ELISA and symptoms were assessed using questionnaires. Evaluation of maternal and infant symptomatology revealed that infected mothers reported more symptoms than vaccinated mothers. We found that after vaccination, human-milk anti-SARS-CoV-2 antibodies persisted for up to 8 months. In addition, distinct patterns of human milk IgA and IgG production we observed after breakthrough infection compared to 3-dose vaccination series alone, indicating a differential central and mucosal immune profiles in hybrid compared with vaccine-induced immunity. To investigate passively-derived milk antibody protection in infants, we examined the persistence of these antibodies in infant saliva after breastfeeding. We found that IgA was more abundant in infant saliva compared to IgG and persist in infant saliva longer after feeding. Our results delineate the differences in milk antibody response to vaccination as compared to breakthrough infection and emphasize the importance of improving the secretion of IgA antibodies to human milk after vaccination to improve the protection of breastfeeding infants.
Collapse
Affiliation(s)
- Yarden Golan
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, and Institute for Human Genetics, University of California, San Francisco, California, United States of America
| | - Mikias Ilala
- Department of Pediatrics, University of California, San Francisco, California, United States of America
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California, United States of America
| | - Caryl Gay
- Department of Family Health Care Nursing, University of California, San Francisco, California, United States of America
| | - Soumya Hunagund
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, California, United States of America
| | - Christine Y. Lin
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, California, United States of America
| | - Arianna G. Cassidy
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, California, United States of America
| | - Unurzul Jigmeddagva
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, California, United States of America
| | - Lin Li
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, California, United States of America
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, California, United States of America
| | - Nida Ozarslan
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, California, United States of America
| | - Ifeyinwa V. Asiodu
- Department of Family Health Care Nursing, University of California, San Francisco, California, United States of America
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, and Institute for Human Genetics, University of California, San Francisco, California, United States of America
| | - Valerie J. Flaherman
- Department of Pediatrics, University of California, San Francisco, California, United States of America
| | - Stephanie L. Gaw
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, California, United States of America
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, California, United States of America
| | - Mary Prahl
- Department of Pediatrics, University of California, San Francisco, California, United States of America
- Division of Pediatric Infectious Diseases and Global Health, University of California, San Francisco, California, United States of America
| |
Collapse
|
39
|
Gabrielli L, Piccirilli G, Petrisli E, Venturoli S, Borgatti EC, Balboni A, Marangoni A, Lazzarotto T. What is the impact of SARS-CoV-2 infection during pregnancy on child immunity? Expert Rev Anti Infect Ther 2022; 21:495-497. [PMID: 36444751 DOI: 10.1080/14787210.2022.2151436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Affiliation(s)
- Liliana Gabrielli
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
| | - Giulia Piccirilli
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
| | - Evangelia Petrisli
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
| | - Simona Venturoli
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
| | - Eva Caterina Borgatti
- Section of Microbiology, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| | - Alice Balboni
- Section of Microbiology, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| | - Antonella Marangoni
- Section of Microbiology, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| | - Tiziana Lazzarotto
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
- Section of Microbiology, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| |
Collapse
|
40
|
Mena-Tudela D, Iglesias-Casas S, Cervera-Gasch A, Andreu-Pejó L, González-Chordá VM, Valero-Chillerón MJ. Breastfeeding and Obstetric Violence during the SARS-CoV-2 Pandemic in Spain: Maternal Perceptions. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:15737. [PMID: 36497811 PMCID: PMC9740849 DOI: 10.3390/ijerph192315737] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/18/2022] [Accepted: 11/24/2022] [Indexed: 06/17/2023]
Abstract
Catalan legislation, a pioneer in Europe, has defined obstetric violence (OV) as "preventing or hindering access to truthful information, necessary for autonomous and informed decision-making". The definition also states that OV can affect physical and mental health, as well as sexual and reproductive health. Some authors have expressed concern about an increase in OV during the SARS-CoV-2 pandemic. During the pandemic, recommendations were also openly offered on the non-establishment and/or early abandonment of breastfeeding without scientific evidence to support them. Experiencing a traumatic childbirth can influence breastfeeding outcomes. Here, we conducted a cross-sectional study using a self-administered online questionnaire. The sample consisted of women who gave birth in Spain between March 2020 and April 2021. The mean age was 34.41 (±4.23) years. Of the women, 73% were employed, 78.2% had a university education, and almost all were Caucasian. Among the subjects, 3.3% were diagnosed with SARS-CoV-2 during pregnancy and 1% were diagnosed during delivery. Some of the women (1.6%) were advised to stop breastfeeding in order to be vaccinated. Women diagnosed with SARS-CoV-2 during delivery (p = 0.048), belonging to a low social class (p = 0.031), with secondary education (p = 0.029), or who suffered obstetric violence (p < 0.001) perceived less support and that the health care providers were less inclined to resolve doubts and difficulties about breastfeeding. Breastfeeding has been significantly challenged during the pandemic. In addition to all the variables to be considered that make breastfeeding support difficult, we now probably need to add SARS-CoV-2 diagnosis and OV.
Collapse
Affiliation(s)
- Desirée Mena-Tudela
- Department of Nursing, Faculty of Health Sciences, Universitat Jaume I, Avda. Sos I Baynat s/n, 12071 Castellón, Spain
| | - Susana Iglesias-Casas
- Department of Obstetrics, Hospital do Salnés, Villgarcía de Aurousa, 36619 Pontevendra, Spain
| | - Agueda Cervera-Gasch
- Department of Nursing, Faculty of Health Sciences, Universitat Jaume I, Avda. Sos I Baynat s/n, 12071 Castellón, Spain
| | - Laura Andreu-Pejó
- Department of Nursing, Faculty of Health Sciences, Universitat Jaume I, Avda. Sos I Baynat s/n, 12071 Castellón, Spain
| | - Victor Manuel González-Chordá
- Department of Nursing, Faculty of Health Sciences, Universitat Jaume I, Avda. Sos I Baynat s/n, 12071 Castellón, Spain
| | - María Jesús Valero-Chillerón
- Department of Nursing, Faculty of Health Sciences, Universitat Jaume I, Avda. Sos I Baynat s/n, 12071 Castellón, Spain
| |
Collapse
|
41
|
Bode L, Bertrand K, Najera JA, Furst A, Honerkamp-Smith G, Shandling AD, Chambers CD, Camerini D, Campo JJ. Characterization of SARS-CoV-2 antibodies in human milk from 21 women with confirmed COVID-19 infection. Pediatr Res 2022; 93:1626-1633. [PMID: 36434204 PMCID: PMC9702863 DOI: 10.1038/s41390-022-02360-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 08/06/2022] [Accepted: 08/27/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND One potential mechanism for protection from SARS-CoV-2 in children is through passive immunity via breast milk from a mother infected with the novel coronavirus. The primary objectives of this study were to establish the presence of SARS-CoV-2-specific IgA and IgG and to characterize the antigenic regions of SARS-CoV-2 proteins that were reactive with antibodies in breast milk. METHODS Between March 2020 and September 2020, 21 women with confirmed SARS-CoV-2 infection were enrolled in Mommy's Milk. Participants donated serial breast milk samples around their time of illness. Breast milk samples were used to probe a multi-coronavirus protein microarray containing full-length and variable-length overlapping fragments of SARS-CoV-2 proteins. Samples were also tested against S and N proteins by electrochemiluminescence assay. RESULTS The breast milk samples contained IgA reactive with a variety of SARS-CoV-2 antigens. The most IgA-reactive SARS-CoV-2 proteins were N (42.9% of women responded to ≥1 N fragment) and S proteins (23.9% responded to ≥1 fragment of S1 or S2). IgG responses were similar. A striking observation was the dissimilarity between mothers in antibody recognition, giving distinct antibody reactivity and kinetic profiles. CONCLUSIONS Individual COVID-19 cases had diverse and unique milk IgA profiles following the onset of symptoms. IMPACT In this observational longitudinal case series of 21 women with confirmed SARS-CoV-2 infection, IgA binding to SARS-CoV-2 proteins detected by orthologous proteome microarray and electrochemiluminescence assays was observed in >75% of women, but there was heterogeneity in which antigens and how many were reactive between women. Immunological profiles of protein regions recognized by each woman were distinct. Diverse repertoires of mucosal breast milk antibody to SARS-CoV-2 reflect heterogeneous passive transfer of maternal antibody to exposed breastfeeding infants.
Collapse
Affiliation(s)
- Lars Bode
- Departments of Pediatrics, University of California, San Diego, La Jolla, CA, 92093, USA. .,Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Kerri Bertrand
- grid.266100.30000 0001 2107 4242Departments of Pediatrics, University of California, San Diego, La Jolla, CA 92093 USA
| | - Julia A. Najera
- grid.266100.30000 0001 2107 4242Departments of Pediatrics, University of California, San Diego, La Jolla, CA 92093 USA ,grid.266100.30000 0001 2107 4242Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California, San Diego, La Jolla, CA 92093 USA
| | - Annalee Furst
- grid.266100.30000 0001 2107 4242Departments of Pediatrics, University of California, San Diego, La Jolla, CA 92093 USA ,grid.266100.30000 0001 2107 4242Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California, San Diego, La Jolla, CA 92093 USA
| | - Gordon Honerkamp-Smith
- grid.266100.30000 0001 2107 4242Departments of Pediatrics, University of California, San Diego, La Jolla, CA 92093 USA
| | | | - Christina D. Chambers
- grid.266100.30000 0001 2107 4242Departments of Pediatrics, University of California, San Diego, La Jolla, CA 92093 USA ,grid.266100.30000 0001 2107 4242Herbert Wertheim School of Public Health and Longevity Science, University of California, San Diego, La Jolla, CA 92093 USA
| | - David Camerini
- grid.420905.aAntigen Discovery Inc. (ADI), Irvine, CA 92618 USA ,grid.266093.80000 0001 0668 7243Center for Virus Research, University of California, Irvine, CA 92697 USA
| | - Joseph J. Campo
- grid.420905.aAntigen Discovery Inc. (ADI), Irvine, CA 92618 USA
| |
Collapse
|
42
|
Lou F, Hu R, Chen Y, Li M, An X, Song L, Tong Y, Fan H. 2'-Fucosyllactose Inhibits Coxsackievirus Class A Type 9 Infection by Blocking Virus Attachment and Internalisation. Int J Mol Sci 2022; 23:ijms232213727. [PMID: 36430203 PMCID: PMC9691179 DOI: 10.3390/ijms232213727] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/18/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022] Open
Abstract
Coxsackieviruses, a genus of enteroviruses in the small RNA virus family, cause fatal infectious diseases in humans. Thus far, there are no approved drugs to prevent these diseases. Human milk contains various biologically active components against pathogens. Currently, the potential activity of breast milk components against the coxsackievirus remains unclear. In our study, the inhibitory effect of 16 major human milk components was tested on coxsackievirus class A type 9 isolate (CV-A9), BUCT01; 2'-Fucosyllactose (2'-FL) was identified to be effective. Time-of-addition, attachment internalisation assays, and the addition of 2'-FL at different time points were applied to investigate its specific role in the viral life cycle. Molecular docking was used to predict 2'-FL's specific cellular targets. The initial screening revealed a significant inhibitory effect (99.97%) against CV-A9 with 10 mg/mL 2'-FL, with no cytotoxicity observed. Compared with the control group, 2'-FL blocked virus entry (85%) as well as inhibited viral attachment (48.4%) and internalisation (51.3%), minimising its infection in rhabdomyosarcoma (RD) cells. The cell pre-incubation with 2'-FL exhibited significant inhibition (73.2-99.9%). Extended incubation between cells with 2'-FL reduced CV-A9 infection (93.9%), suggesting that 2'-FL predominantly targets cells to block infection. Molecular docking results revealed that 2'-FL interacted with the attachment receptor αvβ6 and the internalisation receptor FCGRT and β2M with an affinity of -2.14, -1.87, and -5.43 kcal/mol, respectively. This study lays the foundation for using 2'-FL as a food additive against CV-A9 infections.
Collapse
|
43
|
Alfaro Blazquez R, González-Timoneda A, González-Timoneda M, Gómez Gómez M, Borrull-Guardeño J. Breastfeeding Women's Attitudes About the SARS-COV-2 Vaccine in Spain. J Hum Lact 2022; 38:609-618. [PMID: 35848166 DOI: 10.1177/08903344221109592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Severe Acute Respiratory Syndrome Coronavirus 2 has spread globally, causing the coronavirus disease 2019 (COVID-19) pandemic. To control the pandemic and to achieve herd immunity, four vaccines have obtained market authorization in Europe. Researchers have reported that the sequence of administration of the vaccines depends on the risk of exposure to COVID-19 and age, recommending the vaccine to pregnant and breastfeeding women. RESEARCH AIM To examine the knowledge, willingness and attitudes of breastfeeding women in Spain regarding the possibility of receiving the COVID-19 vaccine. METHODS An observational descriptive cross-sectional study in two tertiary-level hospitals from Valencia was conducted. Breastfeeding women (N = 301) were recruited by non-probability sampling of consecutive cases. Questionnaires were collected in June 2021. RESULTS More than one in every two participants would accept a COVID-19 vaccine, and the percentage rose in participants who were older, more educated, or worked in high-risk jobs. Their main source of information was the Internet, and midwives were the professionals who most recommended vaccination. Participants who would not accept vaccination during lactation reported reasons linked to lack of information regarding vaccination in their condition and how it might affect newborns. CONCLUSION It is of primary concern for healthcare professionals to foster a greater understanding by providing updated information regarding the need, safety, and efficacy of the vaccine for both lactating mothers and their newborns.
Collapse
Affiliation(s)
- Ruben Alfaro Blazquez
- Department of Obstetrics and Gynaecology, University and Polytechnic Hospital "La Fe", Valencia, Spain.,Faculty of Nursing and Podiatry, University of Valencia, Valencia, Spain
| | - Alba González-Timoneda
- Department of Obstetrics and Gynaecology, University and Polytechnic Hospital "La Fe", Valencia, Spain.,Faculty of Nursing and Podiatry, University of Valencia, Valencia, Spain.,Faculty of Nursing, European University of Valencia, Valencia, Spain
| | | | - María Gómez Gómez
- Department of Obstetrics and Gynaecology, University and Polytechnic Hospital "La Fe", Valencia, Spain
| | | |
Collapse
|
44
|
Martin MA, Keith M, Pace RM, Williams JE, Ley SH, Barbosa-Leiker C, Caffé B, Smith CB, Kunkle A, Lackey KA, Navarrete AD, Pace CDW, Gogel AC, Eisenberg DT, Fehrenkamp BD, McGuire MA, McGuire MK, Meehan CL, Brindle E. SARS-CoV-2 specific antibody trajectories in mothers and infants over two months following maternal infection. Front Immunol 2022; 13:1015002. [PMID: 36304449 PMCID: PMC9596287 DOI: 10.3389/fimmu.2022.1015002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/22/2022] [Indexed: 12/15/2022] Open
Abstract
Infants exposed to caregivers infected with SARS-CoV-2 may have heightened infection risks relative to older children due to their more intensive care and feeding needs. However, there has been limited research on COVID-19 outcomes in exposed infants beyond the neonatal period. Between June 2020 - March 2021, we conducted interviews and collected capillary dried blood spots from 46 SARS-CoV-2 infected mothers and their infants (aged 1-36 months) for up to two months following maternal infection onset (COVID+ group, 87% breastfeeding). Comparative data were also collected from 26 breastfeeding mothers with no known SARS-CoV-2 infection or exposures (breastfeeding control group), and 11 mothers who tested SARS-CoV-2 negative after experiencing symptoms or close contact exposure (COVID- group, 73% breastfeeding). Dried blood spots were assayed for anti-SARS-CoV-2 S-RBD IgG and IgA positivity and anti-SARS-CoV-2 S1 + S2 IgG concentrations. Within the COVID+ group, the mean probability of seropositivity among infant samples was lower than that of corresponding maternal samples (0.54 and 0.87, respectively, for IgG; 0.33 and 0.85, respectively, for IgA), with likelihood of infant infection positively associated with the number of maternal symptoms and other household infections reported. COVID+ mothers reported a lower incidence of COVID-19 symptoms among their infants as compared to themselves and other household adults, and infants had similar PCR positivity rates as other household children. No samples returned by COVID- mothers or their infants tested antibody positive. Among the breastfeeding control group, 44% of mothers but none of their infants tested antibody positive in at least one sample. Results support previous research demonstrating minimal risks to infants following maternal COVID-19 infection, including for breastfeeding infants.
Collapse
Affiliation(s)
- Melanie A. Martin
- Department of Anthropology, University of Washington, Seattle, WA, United States,Center for Studies in Demography and Ecology, University of Washington, Seattle, WA, United States,*Correspondence: Melanie A. Martin,
| | - Monica Keith
- Department of Anthropology, University of Washington, Seattle, WA, United States,Center for Studies in Demography and Ecology, University of Washington, Seattle, WA, United States
| | - Ryan M. Pace
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Janet E. Williams
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID, United States
| | - Sylvia H. Ley
- Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA, United States
| | - Celestina Barbosa-Leiker
- College of Nursing, Washington State University Health Sciences Spokane, Spokane, WA, United States
| | - Beatrice Caffé
- Department of Anthropology, Washington State University, Pullman, WA, United States
| | - Caroline B. Smith
- Department of Anthropology, Washington State University, Pullman, WA, United States
| | - Amanda Kunkle
- Department of Anthropology, University of Washington, Seattle, WA, United States
| | - Kimberly A. Lackey
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Alexandra D. Navarrete
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Christina D. W. Pace
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Alexandra C. Gogel
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Dan T.A. Eisenberg
- Department of Anthropology, University of Washington, Seattle, WA, United States,Center for Studies in Demography and Ecology, University of Washington, Seattle, WA, United States
| | - Bethaney D. Fehrenkamp
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States,Washington, Wyoming, Alaska, Montana and Idaho (WWAMI) Medical Education, University of Idaho, Moscow, ID, United States
| | - Mark A. McGuire
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID, United States
| | - Michelle K. McGuire
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Courtney L. Meehan
- Department of Anthropology, Washington State University, Pullman, WA, United States
| | - Eleanor Brindle
- Center for Studies in Demography and Ecology, University of Washington, Seattle, WA, United States,Maternal, Newborn and Child Health & Nutrition, PATH, Seattle, WA, United States
| |
Collapse
|
45
|
Longueira Y, Ojeda DS, Battistelli RBA, Sanchez L, Oviedo Rouco S, Albano D, Guevara E, Valls V, Pando MA, Gamarnik AV. SARS-CoV-2-Specific IgG and IgA response in maternal blood and breastmilk of vaccinated naïve and convalescent lactating participants. Front Immunol 2022; 13:909995. [PMID: 36263055 PMCID: PMC9574440 DOI: 10.3389/fimmu.2022.909995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 09/01/2022] [Indexed: 12/31/2022] Open
Abstract
Background Recent studies have shown the presence of SARS-CoV-2-specific antibodies in the milk of breastfeeding mothers vaccinated with mRNA and convalescent. However, limited information is available in lactating women receiving other vaccine platforms used in developing countries, such as the inactivated SARS-CoV-2 vaccine BBIBP-CorV (Sinopharm) and the non-replicating adenovirus vaccines Sputnik V (Gamaleya Institute) and ChAdOx1-S (Oxford AstraZeneca). Methods Here, we evaluated anti-SARS-CoV-2 IgG and IgA levels in both serum and milk samples using a longitudinal and a cross-sectional cohort of 208 breastfeeding vaccinated women from Argentina with or without previous SARS-CoV-2 infection. Results The analysis showed that IgA levels remain constant in serum and milk of breastfeeding mothers between the first and second doses of vector-based vaccines (Sputnik V and ChAdOx1-S). After the second dose, anti-spike IgA was found positive in 100% of the serum samples and in 66% of breastmilk samples. In addition, no significant differences in milk IgA levels were observed in participants receiving BBIBP-CorV, Sputnik V or ChAdOx1-S. IgG levels in milk increased after the second dose of vector-based vaccines. Paired longitudinal samples taken at 45 and 120 days after the second dose showed a decrease in milk IgG levels over time. Study of IgA levels in serum and milk of vaccinated naïve of infection and vaccinated-convalescent breastfeeding participants showed significantly higher levels in vaccinated-convalescent than in participants without previous infection. Conclusion This study is relevant to understand the protection against SARS-CoV-2 by passive immunity in newborns and children who are not yet eligible to receive vaccination.
Collapse
Affiliation(s)
- Yesica Longueira
- CONICET – Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina
| | - Diego S. Ojeda
- Laboratorio de Virología Molecular, Fundación Instituto Leloir-CONICET, Buenos Aires, Argentina,Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología, Buenos Aires, Argentina,*Correspondence: Andrea V. Gamarnik, ; Diego S. Ojeda,
| | - Rocio B. Antivero Battistelli
- CONICET – Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina,Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología, Buenos Aires, Argentina
| | - Lautaro Sanchez
- Laboratorio de Virología Molecular, Fundación Instituto Leloir-CONICET, Buenos Aires, Argentina
| | - Santiago Oviedo Rouco
- Laboratorio de Virología Molecular, Fundación Instituto Leloir-CONICET, Buenos Aires, Argentina
| | - Daniel Albano
- Banco de Leche Humana – Hospital Materno Infantil Ramón Sardá, Ciudad Autónoma de Buenos Aires, Argentina
| | - Eleonora Guevara
- Banco de Leche Humana – Hospital Materno Infantil Ramón Sardá, Ciudad Autónoma de Buenos Aires, Argentina
| | - Vanesa Valls
- Banco de Leche Humana – Hospital Materno Infantil Ramón Sardá, Ciudad Autónoma de Buenos Aires, Argentina
| | - María A. Pando
- CONICET – Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina,Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología, Buenos Aires, Argentina
| | - Andrea V. Gamarnik
- Laboratorio de Virología Molecular, Fundación Instituto Leloir-CONICET, Buenos Aires, Argentina,*Correspondence: Andrea V. Gamarnik, ; Diego S. Ojeda,
| |
Collapse
|
46
|
Krogstad P, Contreras D, Ng H, Tobin N, Chambers CD, Bertrand K, Bode L, Aldrovandi GM. No infectious SARS-CoV-2 in breast milk from a cohort of 110 lactating women. Pediatr Res 2022; 92:1140-1145. [PMID: 35042956 PMCID: PMC9586866 DOI: 10.1038/s41390-021-01902-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/30/2021] [Accepted: 09/17/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND Genomic RNA of severe acute respiratory syndrome-associated coronavirus type 2 (SARS-CoV-2) has been detected in the breast milk of lactating women, but its pathological significance has remained uncertain due to the small size of prior studies. METHODS Breast milk from 110 lactating women was analyzed by reverse transcription-polymerase chain reaction (285 samples) and viral culture (160 samples). Those containing SARS-CoV-2 viral RNA (vRNA) were examined for the presence of subgenomic RNA (sgRNA), a putative marker of infectivity. RESULTS Sixty-five women had a positive SARS-CoV-2 diagnostic test, 9 had symptoms but negative diagnostic tests, and 36 symptomatic women were not tested. SARS-CoV-2 vRNA was detected in the milk of 7 (6%) women with either a confirmed infection or symptomatic illness, including 6 of 65 (9%) women with a positive SARS-CoV-2 diagnostic test. Infectious virus was not detected in any culture and none had detectable sgRNA. In control experiments, infectious SARS-CoV-2 could be cultured after addition to breastmilk despite several freeze-thaw cycles, as it occurs in the storage and usage of human milk. CONCLUSIONS SARS-CoV-2 RNA can be found infrequently in the breastmilk after recent infection, but we found no evidence that breastmilk contains an infectious virus or that breastfeeding represents a risk factor for transmission of infection to infants. IMPACT This article goes beyond prior small studies to provide evidence that infectious SARS-CoV-2 is not present in the milk of lactating women with recent infection, even when SARS-CoV-2 RNA is detected. Recent SARS-CoV-2 infection or detection of its RNA in human milk is not a contraindication to breastfeeding.
Collapse
Affiliation(s)
- Paul Krogstad
- Department of Pediatrics, David Geffen Scool of Medicine at UCLAs, University of California, Los Angeles, CA, USA.
- Departments of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, USA.
| | - Deisy Contreras
- Department of Pediatrics, David Geffen Scool of Medicine at UCLAs, University of California, Los Angeles, CA, USA
| | - Hwee Ng
- Department of Pediatrics, David Geffen Scool of Medicine at UCLAs, University of California, Los Angeles, CA, USA
| | - Nicole Tobin
- Department of Pediatrics, David Geffen Scool of Medicine at UCLAs, University of California, Los Angeles, CA, USA
| | - Christina D Chambers
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
- Hebert Wertheim School of Public Health and Longevity Science, University of California, San Diego, La Jolla, CA, USA
| | - Kerri Bertrand
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
- Hebert Wertheim School of Public Health and Longevity Science, University of California, San Diego, La Jolla, CA, USA
| | - Lars Bode
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
- Larsson-RosenquistFoundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California, San Diego, La Jolla, CA, USA
| | - Grace M Aldrovandi
- Department of Pediatrics, David Geffen Scool of Medicine at UCLAs, University of California, Los Angeles, CA, USA
| |
Collapse
|
47
|
Gu Y, Low JM, Tan JSY, Ng MSF, Ng LFP, Shunmuganathan B, Gupta R, MacAry PA, Amin Z, Lee LY, Lian D, Shek LPC, Zhong Y, Wang LW. Immune and pathophysiologic profiling of antenatal coronavirus disease 2019 in the GIFT cohort: A Singaporean case-control study. Front Pediatr 2022; 10:949756. [PMID: 36186648 PMCID: PMC9521552 DOI: 10.3389/fped.2022.949756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
COVID-19 can be severe in pregnant women, and have adverse consequences for the subsequent infant. We profiled the post-infectious immune responses in maternal and child blood as well as breast milk in terms of antibody and cytokine expression and performed histopathological studies on placentae obtained from mothers convalescent from antenatal COVID-19. Seventeen mother-child dyads (8 cases of antenatal COVID-19 and 9 healthy unrelated controls; 34 individuals in total) were recruited to the Gestational Immunity For Transfer (GIFT) study. Maternal and infant blood, and breast milk samples were collected over the first year of life. All samples were analyzed for IgG and IgA against whole SARS-CoV-2 spike protein, the spike receptor-binding domain (RBD), and previously reported immunodominant epitopes, as well as cytokine levels. The placentae were examined microscopically. The study is registered at clinicaltrials.gov under the identifier NCT04802278. We found high levels of virus-specific IgG in convalescent mothers and similarly elevated titers in newborn children. Thus, antenatal SARS-CoV-2 infection led to high plasma titers of virus-specific antibodies in infants postnatally. However, this waned within 3-6 months of life. Virus neutralization by plasma was not uniformly achieved, and the presence of antibodies targeting known immunodominant epitopes did not assure neutralization. Virus-specific IgA levels were variable among convalescent individuals' sera and breast milk. Antibody transfer ratios and the decay of transplacentally transferred virus-specific antibodies in neonatal circulation resembled that for other pathogens. Convalescent mothers showed signs of chronic inflammation marked by persistently elevated IL17RA levels in their blood. Four placentae presented signs of acute inflammation, particularly in the subchorionic region, marked by neutrophil infiltration even though > 50 days had elapsed between virus clearance and delivery. Administration of a single dose of BNT162b2 mRNA vaccine to mothers convalescent from antenatal COVID-19 increased virus-specific IgG and IgA titers in breast milk, highlighting the importance of receiving the vaccine even after natural infection with the added benefit of enhanced passive immunity.
Collapse
Affiliation(s)
- Yue Gu
- Antibody Engineering Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jia Ming Low
- Department of Neonatology, Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, Singapore, Singapore
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Melissa Shu Feng Ng
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Lisa F. P. Ng
- Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore, Singapore
| | - Bhuvaneshwari Shunmuganathan
- Antibody Engineering Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Rashi Gupta
- Antibody Engineering Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Paul A. MacAry
- Antibody Engineering Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Zubair Amin
- Department of Neonatology, Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, Singapore, Singapore
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Le Ye Lee
- Department of Neonatology, Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, Singapore, Singapore
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Derrick Lian
- Department of Pathology, National University Hospital, Singapore, Singapore
| | - Lynette Pei-Chi Shek
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, Singapore, Singapore
| | - Youjia Zhong
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, Singapore, Singapore
| | - Liang Wei Wang
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| |
Collapse
|
48
|
Vertical anti-SARS-CoV-2 monoclonal antibody transfer from mothers to HIV-exposed and unexposed infants. AIDS 2022; 36:1613-1615. [PMID: 35979836 DOI: 10.1097/qad.0000000000003306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
|
49
|
Davis EC, Castagna VP, Sela DA, Hillard MA, Lindberg S, Mantis NJ, Seppo AE, Järvinen KM. Gut microbiome and breast-feeding: Implications for early immune development. J Allergy Clin Immunol 2022; 150:523-534. [PMID: 36075638 PMCID: PMC9463492 DOI: 10.1016/j.jaci.2022.07.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022]
Abstract
Establishment of the gut microbiome during early life is a complex process with lasting implications for an individual's health. Several factors influence microbial assembly; however, breast-feeding is recognized as one of the most influential drivers of gut microbiome composition during infancy, with potential implications for function. Differences in gut microbial communities between breast-fed and formula-fed infants have been consistently observed and are hypothesized to partially mediate the relationships between breast-feeding and decreased risk for numerous communicable and noncommunicable diseases in early life. Despite decades of research on the gut microbiome of breast-fed infants, there are large scientific gaps in understanding how human milk has evolved to support microbial and immune development. This review will summarize the evidence on how breast-feeding broadly affects the composition and function of the early-life gut microbiome and discuss mechanisms by which specific human milk components shape intestinal bacterial colonization, succession, and function.
Collapse
Affiliation(s)
- Erin C Davis
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY
| | | | - David A Sela
- Department of Food Science, University of Massachusetts Amherst, Amherst, Mass; Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Mass; Organismic and Evolutionary Biology Graduate Program, University of Massachusetts Amherst, Amherst, Mass
| | - Margaret A Hillard
- Department of Food Science, University of Massachusetts Amherst, Amherst, Mass; Organismic and Evolutionary Biology Graduate Program, University of Massachusetts Amherst, Amherst, Mass
| | - Samantha Lindberg
- Department of Biomedical Sciences, University of Albany, Rensselaer, NY
| | - Nicholas J Mantis
- Division of Infectious Diseases, New York State Department of Health, Albany, NY
| | - Antti E Seppo
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY
| | - Kirsi M Järvinen
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY; Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY; Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY.
| |
Collapse
|
50
|
Ricciardi A, Zelini P, Cassaniti I, Avanzini MA, Colaneri M, De Silvestri A, Baldanti F, Bruno R. Serum and breastmilk SARS-CoV-2 specific antibodies following BNT162b2 vaccine: prolonged protection from SARS-CoV-2 in newborns and older children. Int J Infect Dis 2022; 122:905-909. [PMID: 35803470 PMCID: PMC9254442 DOI: 10.1016/j.ijid.2022.06.055] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVES Vaccination is the best strategy against COVID-19. We aimed to determine antibodies against SARS-CoV-2 in breastmilk and serum of mothers vaccinated with the mRNA vaccine. METHODS This prospective study included 18 lactating women vaccinated with the BNT162b2 vaccine. Serum and breastmilk were collected before the first dose (T0), at the second dose (T1), 3 weeks after the second dose (T2), and 6 months after the first dose (T3). Serum anti-SARS-CoV-2 Spike (S) Immunoglobulin G (IgG) and Immunoglobulin A (IgA) were measured using a semi-quantitative enzyme-linked immunosorbent assay (ELISA) and secretory antibody (s) IgG and IgA in breastmilk using quantitative analysis. RESULTS We detected serum anti-S IgG and IgA in all women after vaccination. Specific IgG and IgA were higher at T1, T2, and T3 compared with T0 (P <0.0001). Higher antibody levels were observed at T2 and lower values at T3 versus T2 (P = 0.007). After 6 months, all patients had serum IgG, but three of 18 (16%) had serum IgA. In breastmilk, sIgA was present at T1 and T2 and decreased after 6 months at T3 (P = 0.002). Breastmilk sIgG levels increased at T1 and T2 and peaked at T3 (P = 0.008). CONCLUSION Secretory antibodies were transmitted through breastmilk until 6 months after anti-COVID-19 mRNA vaccination. Protection of the newborn through breastfeeding needs to be addressed.
Collapse
Affiliation(s)
- Alessandra Ricciardi
- Department of Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| | - Paola Zelini
- Department of Obstetrics and Gynecology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Irene Cassaniti
- Molecular Virology Unit, Department of Microbiology and Virology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Maria Antonietta Avanzini
- Cell Factory, Pediatric Hematology Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marta Colaneri
- Department of Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Annalisa De Silvestri
- Service of Biometry and Statistics Fondazione IRCCS Policlinico San Matteo, Viale Camillo Golgi 19, 27100, Pavia, Italy
| | - Fausto Baldanti
- Molecular Virology Unit, Department of Microbiology and Virology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Raffaele Bruno
- Department of Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|