1
|
Verhasselt V, Tellier J, Carsetti R, Tepekule B. Antibodies in breast milk: Pro-bodies designed for healthy newborn development. Immunol Rev 2024; 328:192-204. [PMID: 39435770 DOI: 10.1111/imr.13411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
This manuscript sheds light on the impact of maternal breast milk antibodies on infant health. Milk antibodies prepare and protect the newborn against environmental exposure, guide and regulate the offspring's immune system, and promote transgenerational adaptation of the immune system to its environment. While the transfer of IgG across the placenta ceases at birth, milk antibodies are continuously replenished by the maternal immune system. They reflect the mother's real-time adaptation to the environment to which the infant is exposed. They cover the infant's upper respiratory and digestive mucosa and are perfectly positioned to control responses to environmental antigens and might also reach their circulation. Maternal antibodies in breast milk play a key role in the immune defense of the developing child, with a major impact on infectious disease susceptibility in both HIC and LMIC. They also influence the development of another major health burden in children-allergies. Finally, emerging evidence shows that milk antibodies also actively shape immune development. Much of this is likely to be mediated by their effect on the seeding, composition and function of the microbiota, but not only. Further understanding of the bridge that maternal antibodies provide between the child and its environment should enable the best interventions to promote healthy development.
Collapse
Affiliation(s)
- Valerie Verhasselt
- Larsson-Rosenquist Foundation Centre for Immunology and Breastfeeding, School of Medicine and of BioMedical Sciences, University of Western Australia, Perth, Western Australia, Australia
- Immunology and Breastfeeding team, The Kids Research Institute Australia, Perth, Western Australia, Australia
| | - Julie Tellier
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Rita Carsetti
- B-cell Lab, Bambino Gesù Children Hospital, Rome, Italy
| | - Burcu Tepekule
- Dept of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, USA
| |
Collapse
|
2
|
Stolojanu C, Doros G, Bratu ML, Ciobanu I, Munteanu K, Iacob ER, Ghenciu LA, Stoicescu ER, Dima M. COVID-19 and Its Potential Impact on Children Born to Mothers Infected During Pregnancy: A Comprehensive Review. Diagnostics (Basel) 2024; 14:2443. [PMID: 39518410 PMCID: PMC11545714 DOI: 10.3390/diagnostics14212443] [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: 09/19/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Pregnancy is a vulnerable period of time during which pregnant people are prone to infections like COVID-19, which can increase risks for both the mother and fetus. These infections may lead to complications such as preterm birth, developmental delays, and congenital abnormalities. While COVID-19 poses additional risks like placental dysfunction and neonatal infections, studies on long-term effects remain limited. Ongoing research and monitoring are essential to understand and mitigate potential cognitive and developmental challenges in children born to mothers infected with COVID-19. This review aims to guide clinicians in managing these risks throughout childhood. Maternal COVID-19 infection during pregnancy can have significant implications for fetal development, even if the newborn is not infected at birth. The release of inflammatory cytokines may cross the placental barrier, potentially disrupting fetal brain development and increasing the risk of long-term cognitive and behavioral issues, such as ADHD or autism. Placental dysfunction, caused by inflammation or thrombosis, can lead to intrauterine growth restriction (IUGR), preterm birth, or hypoxia, affecting both neurological and respiratory health in newborns. Furthermore, a compromised fetal immune system can increase susceptibility to autoimmune conditions and infections. The early diagnosis and management of infections during pregnancy are crucial in mitigating risks to both the mother and fetus. Swift intervention can prevent complications like preterm birth and long-term developmental challenges, ensuring better health outcomes for both the mother and child. Long-term monitoring of children born to mothers infected with COVID-19 is necessary to understand the full extent of the virus's impact. This review evaluates the long-term systemic effects of maternal COVID-19 infection during pregnancy on fetuses, newborns, and children, focusing beyond vertical transmission. It highlights the broader impacts on fetal development, offering insights to help clinicians manage potential issues that may arise later in life.
Collapse
Affiliation(s)
- Cristiana Stolojanu
- Doctoral School, ‘Victor Babes’ University of Medicine and Pharmacy, 300041 Timisoara, Romania;
- ‘Louis Turcanu’ Emergency Hospital for Children, 300011 Timisoara, Romania;
| | - Gabriela Doros
- ‘Louis Turcanu’ Emergency Hospital for Children, 300011 Timisoara, Romania;
- Department of Pediatrics, ‘Victor Babes’ University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Melania Lavinia Bratu
- Center for Neuropsychology and Behavioral Medicine, Discipline of Psychology, Faculty of General Medicine, ‘Victor Babes’ University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
- Center for Cognitive Research in Neuropsychiatric Pathology, Department of Neurosciences, ‘Victor Babes’ University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
| | - Iulia Ciobanu
- Department of Anatomy and Embriology, ‘Victor Babes’ Univeristy of Medicine and Pharmacy, 300041 Timisoara, Romania; (I.C.); (K.M.)
| | - Krisztina Munteanu
- Department of Anatomy and Embriology, ‘Victor Babes’ Univeristy of Medicine and Pharmacy, 300041 Timisoara, Romania; (I.C.); (K.M.)
| | - Emil Radu Iacob
- Department of Pediatric Surgery, ‘Victor Babes’ University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Laura Andreea Ghenciu
- Department of Functional Sciences, ‘Victor Babes’ University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania;
| | - Emil Robert Stoicescu
- Radiology and Medical Imaging University Clinic, ‘Victor Babes’ University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania;
- Research Center for Pharmaco-Toxicological Evaluations, ‘Victor Babes’ University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
- Research Center for Medical Communication, ‘Victor Babes’ University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
- Field of Applied Engineering Sciences, Specialization Statistical Methods and Techniques in Health and Clinical Research, Faculty of Mechanics, ‘Politehnica’ University Timisoara, 300222 Timisoara, Romania
| | - Mirabela Dima
- Department of Neonatology, ‘Victor Babes’ University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| |
Collapse
|
3
|
Bui MT, Nguyen Le CA, Duong KL, Hoang VT, Nguyen TK. Transplacental Transmission of SARS-CoV-2: A Narrative Review. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1517. [PMID: 39336558 PMCID: PMC11434576 DOI: 10.3390/medicina60091517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/12/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024]
Abstract
Background and Objectives: The study aims to explore the potential for transplacental transmission of SARS-CoV-2, focusing on its pathophysiology, placental defense mechanisms, and the clinical implications for maternal and neonatal health. Materials and Methods: A comprehensive review of the current literature was conducted, analyzing studies on SARS-CoV-2 infection in pregnancy, the expression of key viral receptors (ACE2 and TMPRSS2) in placental cells, and the immune responses involved in placental defense. The review also examined the clinical outcomes related to maternal and neonatal health, including adverse pregnancy outcomes and neonatal infection. Results: The expression of ACE2 and TMPRSS2 in the placenta supports the biological plausibility of SARS-CoV-2 transplacental transmission. Histopathological findings from the infected placentas reveal inflammation, vascular changes, and the evidence of viral particles in placental tissues. Clinical reports indicate an increased risk of preterm birth, intrauterine growth restriction, and neonatal infection in pregnancies affected by COVID-19. However, the frequency and mechanisms of vertical transmission remain variable across studies, highlighting the need for standardized research protocols. Conclusions: SARS-CoV-2 can potentially infect placental cells, leading to adverse pregnancy outcomes and neonatal infection. While evidence of transplacental transmission has been documented, the risk and mechanisms are not fully understood. Ongoing research is essential to clarify these aspects and inform obstetric care practices to improve maternal and neonatal outcomes during the COVID-19 pandemic.
Collapse
Affiliation(s)
| | | | | | | | - Trung Kien Nguyen
- Thai Binh University of Medicine and Pharmacy, Thai Binh 410000, Vietnam; (M.T.B.); (C.A.N.L.); (K.L.D.); (V.T.H.)
| |
Collapse
|
4
|
Dangi T, Sanchez S, Awakoaiye B, Lew MH, Irani N, Penaloza-MacMaster P. Breast Milk-Derived Antibodies Impair Vaccine Immunity in Suckling Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:612-618. [PMID: 39007643 PMCID: PMC11333162 DOI: 10.4049/jimmunol.2400277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024]
Abstract
Breast milk confers multiple benefits to the neonate, including passive immunity against multiple microorganisms via Abs. However, it remains unclear whether breast milk-derived Abs affect vaccine-induced immunity in the neonate. We evaluated in C57BL/6 and BALB/c mice whether breastfeeding from an mRNA-SARS-CoV-2-vaccinated dam affects vaccine-induced immunity in neonate mice. Using an experimental model that allows the distinction of maternal Abs and neonate Abs based on their allotype, we show that breastfeeding from an immune dam is associated with reduced vaccine immunity in the neonate. Importantly, mice that breastfed from an immune dam showed reduced numbers of plasma cells after vaccination, relative to mice that breastfed from a naive dam. Our subsequent studies using an mRNA-luciferase reporter system show that passive transfer of Abs through breastfeeding accelerates the clearance of vaccine Ag in suckling mice, resulting in reduced Ag availability. Altogether, maternal Abs transferred through breast milk can protect against infectious microorganisms, but they may also interfere with the neonate's response to vaccination by accelerating the clearance of vaccine Ag. These findings are important for understanding the effects of maternal Abs on the neonate's response to vaccines and may provide insights for improving neonatal vaccines.
Collapse
Affiliation(s)
- Tanushree Dangi
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Sarah Sanchez
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Bakare Awakoaiye
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Min Han Lew
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Nahid Irani
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Pablo Penaloza-MacMaster
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
5
|
Verhasselt V. A newborn's perspective on immune responses to food. Immunol Rev 2024; 326:117-129. [PMID: 39162048 DOI: 10.1111/imr.13376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
In this review, we will highlight infants' immune responses to food, emphasizing the unique aspects of early-life immunity and the critical role of breast milk as a food dedicated to infants. Infants are susceptible to inflammatory responses rather than immune tolerance at the mucosal and skin barriers, necessitating strategies to promote oral tolerance that consider this susceptibility. Breast milk provides nutrients for growth and cell metabolism, including immune cells. The content of breast milk, influenced by maternal genetics and environmental exposures, prepares the infant's immune system for the outside world, including solid foods. To do this, breast milk promotes immune system development through antigen-specific and non-antigen-specific immune education by exposing the newborn to food and respiratory allergens and acting on three key targets for food allergy prevention: the gut microbiota, epithelial cells, and immune cells. Building knowledge of how the maternal exposome and human milk composition influence offspring's healthy immune development will lead to recommendations that meet the specific needs of the developing immune system and increase the chances of promoting an appropriate immune response to food in the long term.
Collapse
Affiliation(s)
- Valerie Verhasselt
- Larsson-Rosenquist Foundation Centre for Immunology and Breastfeeding, School of Medicine, University of Western Australia, Perth, Western Australia, Australia
- Immunology and Breastfeeding team, Telethon Kids Institute, Perth, Western Australia, Australia
| |
Collapse
|
6
|
Li L, Matsui Y, Prahl MK, Cassidy AG, Golan Y, Jigmeddagva U, Ozarslan N, Lin CY, Buarpung S, Gonzalez VJ, Chidboy MA, Basilio E, Lynch KL, Song D, Jegatheesan P, Rai DS, Govindaswami B, Needens J, Rincon M, Myatt L, Taha TY, Montano M, Ott M, Greene WC, Gaw SL. Neutralizing and binding antibody responses to SARS-CoV-2 with hybrid immunity in pregnancy. NPJ Vaccines 2024; 9:156. [PMID: 39191763 PMCID: PMC11349990 DOI: 10.1038/s41541-024-00948-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Abstract
Hybrid immunity against SARS-CoV-2 has not been well studied in pregnancy. We conducted a comprehensive analysis of neutralizing antibodies (nAb) and binding antibodies in pregnant individuals who received mRNA vaccination, natural infection, or both. A third vaccine dose augmented nAb levels compared to the two-dose regimen or natural infection alone; this effect was more pronounced in hybrid immunity. There was reduced anti-Omicron nAb, but the maternal-fetal transfer efficiency remained comparable to that of other variants. Vaccine-induced nAbs were transferred more efficiently than infection-induced nAbs. Anti-spike receptor binding domain (RBD) IgG was associated with nAb against wild-type (Wuhan-Hu-1) following breakthrough infection. Both vaccination and infection-induced anti-RBD IgA, which was more durable than anti-nucleocapsid IgA. IgA response was attenuated in pregnancy compared to non-pregnant controls. These data provide additional evidence of augmentation of humoral immune responses in hybrid immunity in pregnancy.
Collapse
Affiliation(s)
- Lin Li
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Yusuke Matsui
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Mary K Prahl
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- Division of Pediatric Infectious Diseases and Global Health, University of California San Francisco, San Francisco, CA, USA
| | - Arianna G Cassidy
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Yarden Golan
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Unurzul Jigmeddagva
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Nida Ozarslan
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Christine Y Lin
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Sirirak Buarpung
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Veronica J Gonzalez
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Megan A Chidboy
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Emilia Basilio
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Kara L Lynch
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Dongli Song
- Division of Neonatology, Department of Pediatrics, Santa Clara Valley Medical Center, San Jose, CA, USA
| | - Priya Jegatheesan
- Division of Neonatology, Department of Pediatrics, Santa Clara Valley Medical Center, San Jose, CA, USA
| | - Daljeet S Rai
- Stanford-O'Connor Family Medicine Residency Program, Division of Family Medicine, Stanford University, Palo Alto, CA, USA
| | - Balaji Govindaswami
- Division of Neonatology, Department of Pediatrics, Marshall University Joan C Edwards School of Medicine, Huntington, WV, USA
| | - Jordan Needens
- Department of Obstetrics and Gynecology, Marshall University Joan C Edwards School of Medicine, Huntington, WV, USA
| | - Monica Rincon
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, USA
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, USA
| | - Taha Y Taha
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Mauricio Montano
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Melanie Ott
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Departments of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Warner C Greene
- Gladstone Institute of Virology, San Francisco, CA, USA.
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA.
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
- Departments of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA.
| | - Stephanie L Gaw
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA.
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
7
|
Öztürk N, Kaya Z, Boyunağa Ö, Söylemezoğlu O. An unusual cause of renal vein thrombosis in a newborn: COVID-19. CEN Case Rep 2024; 13:302-305. [PMID: 38189897 PMCID: PMC11294506 DOI: 10.1007/s13730-023-00846-y] [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: 09/03/2023] [Accepted: 12/01/2023] [Indexed: 01/09/2024] Open
Abstract
There is no information on renal vein thrombosis induced by COVID-19 infection in a neonate. Few cases of renal vein thrombosis caused by COVID-19 infection have been reported in predominantly adult patients. On day 25 after birth, a newborn whose mother was infected with COVID-19 had renal vein thrombosis. We believed that our patient's renal vein thrombosis was caused by postnatal transmission of the COVID-19 infection that the mother had acquired during birth. The clinical and radiologic findings of these unusual renal complications in a neonate, as well as treatment options, are presented.
Collapse
Affiliation(s)
- Nihan Öztürk
- Department of Pediatric Hematology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Zühre Kaya
- Department of Pediatric Hematology, Faculty of Medicine, Gazi University, Ankara, Turkey.
| | - Öznur Boyunağa
- Department of Radiology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Oğuz Söylemezoğlu
- Department of Pediatric Nephrology, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
8
|
Fu X, Du B, Chen PA, Shama A, Chen B, Zhang X, Han X, Xu Y, Gong Y, Zeng X, Sun C, Yang W, Xing X, Li Z, Fu Y, Ke D, Wang N, Xia Y, Sun Y, Chen Q. Exploring the impact of gut microbial metabolites on inactivated SARS-CoV-2 vaccine efficacy during pregnancy and mother-to-infant antibody transfer. Gut 2024; 73:1397-1400. [PMID: 37739779 PMCID: PMC11287518 DOI: 10.1136/gutjnl-2023-330497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/30/2023] [Indexed: 09/24/2023]
Affiliation(s)
- Xi Fu
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Schoold of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Bingqian Du
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Schoold of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Pei-An Chen
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Schoold of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Aga Shama
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Schoold of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Baolan Chen
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Schoold of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Xi Zhang
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Schoold of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Xue Han
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Schoold of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Yingxia Xu
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Schoold of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Yajie Gong
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Schoold of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Xia Zeng
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Schoold of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Chongzhen Sun
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Schoold of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Wenhan Yang
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Schoold of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Xiaohui Xing
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Schoold of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Zhongjun Li
- Dongguan People's Hospital, Dongguan, Guangdong, China
| | - Yanyan Fu
- Guangzhou Baiyun District Maternal and Child Health Hospital, Guangzhou, China
| | - Dongyun Ke
- Guangzhou Baiyun District Maternal and Child Health Hospital, Guangzhou, China
| | - Niping Wang
- Guangzhou Baiyun District Maternal and Child Health Hospital, Guangzhou, China
| | - Yun Xia
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Schoold of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Yu Sun
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, Guangdong, China
| | - Qingsong Chen
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Schoold of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
Jepsen SD, Lund A, Matwiejuk M, Andresen L, Christensen KR, Skov S. Human milk oligosaccharides regulate human macrophage polarization and activation in response to Staphylococcus aureus. Front Immunol 2024; 15:1379042. [PMID: 38903508 PMCID: PMC11187579 DOI: 10.3389/fimmu.2024.1379042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/08/2024] [Indexed: 06/22/2024] Open
Abstract
Human milk oligosaccharides (HMOs) are present in high numbers in milk of lactating women. They are beneficial to gut health and the habitant microbiota, but less is known about their effect on cells from the immune system. In this study, we investigated the direct effect of three structurally different HMOs on human derived macrophages before challenge with Staphylococcus aureus (S. aureus). The study demonstrates that individual HMO structures potently affect the activation, differentiation and development of monocyte-derived macrophages in response to S. aureus. 6´-Sialyllactose (6'SL) had the most pronounced effect on the immune response against S. aureus, as illustrated by altered expression of macrophage surface markers, pointing towards an activated M1-like macrophage-phenotype. Similarly, 6'SL increased production of the pro-inflammatory cytokines TNF-α, IL-6, IL-8, IFN-γ and IL-1β, when exposing cells to 6'SL in combination with S. aureus compared with S. aureus alone. Interestingly, macrophages treated with 6'SL exhibited an altered proliferation profile and increased the production of the classic M1 transcription factor NF-κB. The HMOs also enhanced macrophage phagocytosis and uptake of S. aureus. Importantly, the different HMOs did not notably affect macrophage activation and differentiation without S. aureus exposure. Together, these findings show that HMOs can potently augment the immune response against S. aureus, without causing inflammatory activation in the absence of S. aureus, suggesting that HMOs assist the immune system in targeting important pathogens during early infancy.
Collapse
Affiliation(s)
- Stine Dam Jepsen
- dsm-firmenich, Hørsholm, Denmark
- Immunology, Section for Preclinical Disease Biology, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Astrid Lund
- Immunology, Section for Preclinical Disease Biology, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Lars Andresen
- Immunology, Section for Preclinical Disease Biology, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Søren Skov
- Immunology, Section for Preclinical Disease Biology, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
10
|
Wu YX, Wang DD, Zhao RQ, Jin OX, Yang JY, Zhang MX, Wang LZ. Differences in clinical characteristics of early-onset and late-onset severe acute respiratory syndrome coronavirus 2 infections in neonates. Eur J Pediatr 2024; 183:1741-1750. [PMID: 38233605 PMCID: PMC11001696 DOI: 10.1007/s00431-024-05433-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 01/19/2024]
Abstract
Differences in clinical characteristics of early-onset and late-onset severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections in neonates remain unclear. This study aimed to determine whether there are differences in the main clinical, radiological, and laboratory features of early-onset and late-onset SARS-CoV-2 infections in neonates. This single-center, prospective cohort study enrolled neonates with SARS-CoV-2 infection from December 7, 2022, to January 3, 2023, and evaluated their clinical characteristics during hospitalization. All neonates (N = 58) infected with SARS-CoV-2 within 28 days of birth who were admitted to the neonatal intensive care unit of Taizhou Hospital were included. These neonates were classified into the early-onset (diagnosed within 7 days of birth) and late-onset (diagnosed more than 7 days after birth) groups. The symptoms, treatment, and prognosis of SARS-CoV-2 infection were the main study outcomes. The incidence of hospitalization attributable to SARS-CoV-2 infection was 10.6% (58 of 546 neonates) in Linhai. Sixteen (28%) of the 58 SARS-CoV-2 infections were early-onset cases, and 42 (72%) were late-onset cases. The common symptoms among the late-onset group were fever (p < 0.001) and cough (p < 0.001). Neonates with late-onset SARS-CoV-2 infection (p < 0.001) were significantly more likely to develop pneumonia. Conclusion: The clinical symptoms and rates of pneumonia caused by SARS-CoV-2 infection in neonates differed between the early-onset and late-onset groups. Different clinical management is necessary for neonates with early-onset and late-onset SARS-CoV-2 infections. What is Known: • Neonates are susceptible to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). • Differences in clinical characteristics of early-onset and late-onset SARS-CoV-2 infections in neonates remain unclear. What is New: • Fever and cough were the most common symptoms among neonates with late-onset infection. • Neonates with late-onset SARS-CoV-2 infection were more likely to develop pneumonia.
Collapse
Affiliation(s)
- Yi-Xin Wu
- Department of Pediatrics, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street, Linhai, 317000, Zhejiang, China
| | - Dan-Dan Wang
- Department of Pediatrics, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street, Linhai, 317000, Zhejiang, China
| | - Ru-Qi Zhao
- Department of Pediatrics, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street, Linhai, 317000, Zhejiang, China
| | - Ou-Xuan Jin
- Department of Pediatrics, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street, Linhai, 317000, Zhejiang, China
| | - Jing-Yun Yang
- Department of Pediatrics, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street, Linhai, 317000, Zhejiang, China
| | - Mei-Xian Zhang
- Department of Pediatrics, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street, Linhai, 317000, Zhejiang, China.
- Evidence-based Medicine Center, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, 150 Ximen Street, Linhai, 317000, Zhejiang Province, China.
| | - Li-Zhen Wang
- Department of Pediatrics, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street, Linhai, 317000, Zhejiang, China.
| |
Collapse
|
11
|
Conti MG, Piano Mortari E, Nenna R, Pierangeli A, Sorrentino L, Frasca F, Petrarca L, Mancino E, Di Mattia G, Matera L, Fracella M, Albano C, Scagnolari C, Capponi M, Cinicola B, Carsetti R, Midulla F. SARS-CoV-2-specific mucosal immune response in vaccinated versus infected children. Front Cell Infect Microbiol 2024; 14:1231697. [PMID: 38601739 PMCID: PMC11004290 DOI: 10.3389/fcimb.2024.1231697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 03/05/2024] [Indexed: 04/12/2024] Open
Abstract
The anti-COVID-19 intramuscular vaccination induces a strong systemic but a weak mucosal immune response in adults. Little is known about the mucosal immune response in children infected or vaccinated against SARS-CoV-2. We found that 28% of children had detectable salivary IgA against SARS-CoV-2 even before vaccination, suggesting that, in children, SARS-CoV-2 infection may be undiagnosed. After vaccination, only receptor-binding domain (RBD)-specific IgA1 significantly increased in the saliva. Conversely, infected children had significantly higher salivary RBD-IgA2 compared to IgA1, indicating that infection more than vaccination induces a specific mucosal immune response in children. Future efforts should focus on development of vaccine technologies that also activate mucosal immunity.
Collapse
Affiliation(s)
- Maria Giulia Conti
- Department of Maternal, Child Health and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Eva Piano Mortari
- B Cell Unit, Immunology Research Area, Bambino Gesù Children’s Hospital, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Raffaella Nenna
- Department of Maternal, Child Health and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Alessandra Pierangeli
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Leonardo Sorrentino
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Federica Frasca
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Laura Petrarca
- Department of Maternal, Child Health and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Enrica Mancino
- Department of Maternal, Child Health and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Greta Di Mattia
- Department of Maternal, Child Health and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Luigi Matera
- Department of Maternal, Child Health and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Matteo Fracella
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Christian Albano
- B Cell Unit, Immunology Research Area, Bambino Gesù Children’s Hospital, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Carolina Scagnolari
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Martina Capponi
- Department of Maternal, Child Health and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Bianca Cinicola
- Department of Maternal, Child Health and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Rita Carsetti
- B Cell Unit, Immunology Research Area, Bambino Gesù Children’s Hospital, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Fabio Midulla
- Department of Maternal, Child Health and Urological Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
12
|
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: 2.0] [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
|
13
|
Carrasco Colom J, Manzanares Á, Álvaro Gómez A, Serrano Escribano I, Esquivel E, Pérez-Rivilla A, Moral-Pumarega MT, Aguirre Pascual E, De Vergas J, Reda Del Barrio S, Moraleda C, Epalza C, Fernández-Cooke E, Prieto L, Villaverde S, Zamora B, Herraiz I, Galindo A, Folgueira MD, Delgado R, Blázquez-Gamero D. Clinical outcomes and antibody transfer in a cohort of infants with in utero or perinatal exposure to SARS-CoV-2 (Coronascope Study). Eur J Pediatr 2023; 182:4647-4654. [PMID: 37561198 DOI: 10.1007/s00431-023-05147-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023]
Abstract
We aimed to describe the outcomes, focusing on the hearing and neurological development, of infants born to mothers with COVID-19 during pregnancy and to evaluate the persistence of maternal antibodies in the first months of life. An observational, prospective study at a tertiary hospital in Madrid (Spain) on infants born to mothers with COVID-19 during pregnancy between March and September 2020 was conducted. A follow-up visit at 1-3 months of age with a physical and neurological examination, cranial ultrasound (cUS), SARS-CoV-2 RT-PCR on nasopharyngeal swab, and SARS-CoV-2 serology were performed. Hearing was evaluated at birth through the automated auditory brainstem response and at six months of age through the auditory steady-state response. A neurodevelopmental examination using the Bayley-III scale was performed at 12 months of age. Of 95 infants studied, neurological examination was normal in all of them at the follow-up visit, as was the cUS in 81/85 (95%) infants, with only mild abnormalities in four of them. Serology was positive in 47/95 (50%) infants, which was not associated with symptoms or severity of maternal infection. No hearing loss was detected, and neurodevelopment was normal in 96% of the infants (median Z score: 0). CONCLUSION In this cohort, the majority of infants born to mothers with COVID-19 during pregnancy were healthy infants with a normal cUS, no hearing loss, and normal neurodevelopment in the first year of life. Only half of the infants had a positive serological result during the follow-up. WHAT IS KNOWN • Hearing loss and neurodevelopmental delay in infants born to mothers with COVID-19 during pregnancy has been suggested, although data is inconsistent. Maternal antibody transfer seems to be high, with a rapid decrease during the first weeks of life. WHAT IS NEW • Most infants born to mothers with COVID-19 during pregnancy had normal hearing screening, cranial ultrasound, and neurodevelopmental status at 12 months of life. Antibodies against SARS-CoV-2 were only detected in 50% of the infants at two months of life.
Collapse
Affiliation(s)
- Jaime Carrasco Colom
- Pediatric Infectious Disease Unit, Hospital Universitario 12 de Octubre, Madrid, Spain
- Pediatric Research and Clinical Trials Unit (UPIC), Instituto de Investigación Hospital, RECLIP, 12 de Octubre (imas12), Madrid, Spain
| | - Ángela Manzanares
- Pediatric Infectious Disease Unit, Hospital Universitario 12 de Octubre, Madrid, Spain.
- Pediatric Research and Clinical Trials Unit (UPIC), Instituto de Investigación Hospital, RECLIP, 12 de Octubre (imas12), Madrid, Spain.
| | | | | | - Estrella Esquivel
- Pediatric Research and Clinical Trials Unit (UPIC), Instituto de Investigación Hospital, RECLIP, 12 de Octubre (imas12), Madrid, Spain
| | - Alfredo Pérez-Rivilla
- Microbiology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Maria Teresa Moral-Pumarega
- Neonatology Department, Hospital Universitario 12 de Octubre, RICORS Network, ISCIII, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
| | | | - Joaquín De Vergas
- Pediatric Otorhinolaryngology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Sara Reda Del Barrio
- Pediatric Otorhinolaryngology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Cinta Moraleda
- Pediatric Infectious Disease Unit, Hospital Universitario 12 de Octubre, Madrid, Spain
- Pediatric Research and Clinical Trials Unit (UPIC), Instituto de Investigación Hospital, RECLIP, 12 de Octubre (imas12), Madrid, Spain
| | - Cristina Epalza
- Pediatric Infectious Disease Unit, Hospital Universitario 12 de Octubre, Madrid, Spain
- Pediatric Research and Clinical Trials Unit (UPIC), Instituto de Investigación Hospital, RECLIP, 12 de Octubre (imas12), Madrid, Spain
| | - Elisa Fernández-Cooke
- Pediatric Infectious Disease Unit, Hospital Universitario 12 de Octubre, Madrid, Spain
- Pediatric Research and Clinical Trials Unit (UPIC), Instituto de Investigación Hospital, RECLIP, 12 de Octubre (imas12), Madrid, Spain
| | - Luis Prieto
- Pediatric Infectious Disease Unit, Hospital Universitario 12 de Octubre, Madrid, Spain
- Pediatric Research and Clinical Trials Unit (UPIC), Instituto de Investigación Hospital, RECLIP, 12 de Octubre (imas12), Madrid, Spain
- Universidad Complutense de Madrid, Madrid, Spain
| | - Serena Villaverde
- Pediatric Infectious Disease Unit, Hospital Universitario 12 de Octubre, Madrid, Spain
- Pediatric Research and Clinical Trials Unit (UPIC), Instituto de Investigación Hospital, RECLIP, 12 de Octubre (imas12), Madrid, Spain
| | - Berta Zamora
- Neuropsychology Unit, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Ignacio Herraiz
- Universidad Complutense de Madrid, Madrid, Spain
- Fetal Medicine Unit, Obstetrics and Gynecology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
- Primary Care Interventions to Prevent Chronic Maternal and Child Diseases of Perinatal and Developmental Origin (RICORS Network), Instituto de Salud Carlos III, RD21/0012/0024, Madrid, Spain
| | - Alberto Galindo
- Universidad Complutense de Madrid, Madrid, Spain
- Fetal Medicine Unit, Obstetrics and Gynecology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
- Primary Care Interventions to Prevent Chronic Maternal and Child Diseases of Perinatal and Developmental Origin (RICORS Network), Instituto de Salud Carlos III, RD21/0012/0024, Madrid, Spain
| | - María Dolores Folgueira
- Microbiology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Rafael Delgado
- Microbiology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Daniel Blázquez-Gamero
- Pediatric Infectious Disease Unit, Hospital Universitario 12 de Octubre, Madrid, Spain
- Pediatric Research and Clinical Trials Unit (UPIC), Instituto de Investigación Hospital, RECLIP, 12 de Octubre (imas12), Madrid, Spain
- Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
14
|
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: 0.5] [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
|
15
|
Vigil-Vázquez S, Manzanares Á, Hernanz-Lobo A, Carrasco-García I, Zamora Del Pozo C, Pérez-Pérez A, Rincón-López EM, Santiago-García B, Pintado-Recarte MDP, Alonso-Fernández R, Sánchez-Luna M, Navarro-Gómez ML. Serologic evolution and follow-up to IgG antibodies of infants born to mothers with gestational COVID. BMC Pregnancy Childbirth 2023; 23:623. [PMID: 37648971 PMCID: PMC10469412 DOI: 10.1186/s12884-023-05926-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND It is known that SARS-CoV-2 antibodies from pregnant women with SARS-CoV-2 infection during pregnancy cross the placenta but the duration and the protective effect of these antibodies in infants is scarce. METHODS This prospective study included mothers with SARS-COV-2 infection during pregnancy and their infants from April 2020 to March 2021. IgG antibodies to SARS-CoV-2 spike protein were performed on women and infants at birth and at two and six months during follow-up. Anthropometrical measures and physical and neurological examinations and a clinical history of symptoms and COVID-19 diagnosis were collected. Simple linear regression was performed to compare categorical and continuous variables. To compare the mother's and infant's antibody titers evolution, a mixed linear regression model was used. A predictive model of newborn antibody titers at birth has been established by means of simple stepwise linear regression. RESULTS 51 mother-infant couples were included. 45 (90%) of the mothers and 44 (86.3%) of the newborns had a positive serology al birth. These antibodies were progressively decreasing and were positive in 34 (66.7%) and 7 (13.7%) of infants at 2 and 6 months, respectively. IgG titers of newborns at birth were related to mothers' titers, with a positive moderate correlation (Pearson's correlation coefficient: 0.82, p < 0,001). Fetal/maternal antibodies placental transference rate was 1.3 (IQR: 0.7-2.2). The maternal IgG titers at delivery and the type of maternal infection (acute, recent, or past infection) was significantly related with infants' antibody titers at birth. No other epidemiological or clinical factors were related to antibodies titers. Neurodevelopment, psychomotor development, and growth were normal in 94.2% of infants in the third follow-up visit. No infants had a COVID-19 diagnosis during the follow-up period. CONCLUSIONS Transplacental transfer of maternal antibodies is high in newborns from mothers with recent or past infection at delivery, but these antibodies decrease after the first months of life. Infant's IgG titers were related to maternal IgG titers at delivery. Further studies are needed to learn about the protective role of maternal antibodies in infants.
Collapse
Affiliation(s)
- Sara Vigil-Vázquez
- Department of Neonatology, Hospital General Universitario Gregorio Marañón, Calle O'Donnel 48, Madrid, 28009, Spain.
| | - Ángela Manzanares
- Pediatric Infectious Disease Unit, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Pediatrics Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Alicia Hernanz-Lobo
- Pediatric Infectious Disease Unit, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Pediatrics Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Itziar Carrasco-García
- Pediatric Infectious Disease Unit, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Pediatrics Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Clara Zamora Del Pozo
- Department of Obstetrics and Gynecology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Alba Pérez-Pérez
- Department of Neonatology, Hospital General Universitario Gregorio Marañón, Calle O'Donnel 48, Madrid, 28009, Spain
| | - Elena María Rincón-López
- Pediatric Infectious Disease Unit, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Pediatrics Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Begoña Santiago-García
- Pediatric Infectious Disease Unit, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Pediatrics Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Roberto Alonso-Fernández
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Manuel Sánchez-Luna
- Department of Neonatology, Hospital General Universitario Gregorio Marañón, Calle O'Donnel 48, Madrid, 28009, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - María Luisa Navarro-Gómez
- Pediatric Infectious Disease Unit, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Pediatrics Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
16
|
Chisale MRO, Sinyiza FW, Kaseka PU, Chimbatata CS, Mbakaya BC, Wu TSJ, Nyambalo BW, Chauma-Mwale A, Chilima B, Yu KLJ, Kayira AB. Coronavirus Disease 2019 (COVID-19) Reinfection Rates in Malawi: A Possible Tool to Guide Vaccine Prioritisation and Immunisation Policies. Vaccines (Basel) 2023; 11:1185. [PMID: 37515002 PMCID: PMC10383452 DOI: 10.3390/vaccines11071185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
As the fight against the COVID-19 pandemic continues, reports indicate that the global vaccination rate is still far below the target. Understanding the levels of reinfection may help refocus and inform policymakers on vaccination. This retrospective study in Malawi included individuals and patients who tested for COVID-19 infections via reverse transcriptase polymerase chain reaction (rt-PCR) from the data at the Public Health Institute of Malawi (PHIM). We included all data in the national line list from April 2020 to March 2022. Upon review of 47,032 records, 45,486 were included with a reported 82 (0.18) reinfection representing a rate of 0.55 (95% CI: 0.44-0.68) per 100,000 person-days of follow-up. Most reinfections occurred in the first 90 to 200 days following the initial infection, and the median time to reinfection was 175 days (IQR: 150-314), with a range of 90-563 days. The risk of reinfection was highest in the immediate 3 to 6 months following the initial infection and declined substantially after that, and age demonstrated a significant association with reinfection. Estimating the burden of SARS-CoV-2 reinfections, a specific endurance of the immunity naturally gained, and the role played by risk factors in reinfections is relevant for identifying strategies to prioritise vaccination.
Collapse
Affiliation(s)
- Master R O Chisale
- Faculty of Sciences, Technology and Innovations, Biological Sciences, Mzuzu University, P/Bag 201 Luwinga, Mzuzu, Malawi
- Research Department, Luke International, Mzuzu P.O. Box 1088, Malawi
| | | | - Paul Uchizi Kaseka
- Mzuzu Central Hospital, Ministry of Health, P/Bag 209 Luwinga, Mzuzu, Malawi
| | | | | | - Tsung-Shu Joseph Wu
- Research Department, Luke International, Mzuzu P.O. Box 1088, Malawi
- Overseas Department, Pingtung Christian Hospital, No. 60, Da-lien Rd., Pingtung City 900, Taiwan
| | | | - Annie Chauma-Mwale
- Public Health Institute of Malawi, Ministry of Health, Lilongwe 00265, Malawi
| | - Ben Chilima
- Public Health Institute of Malawi, Ministry of Health, Lilongwe 00265, Malawi
| | - Kwong-Leung Joseph Yu
- Research Department, Luke International, Mzuzu P.O. Box 1088, Malawi
- Overseas Department, Pingtung Christian Hospital, No. 60, Da-lien Rd., Pingtung City 900, Taiwan
| | | |
Collapse
|
17
|
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: 3.0] [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
|
18
|
Chutipongtanate S, Cetinkaya H, Zhang X, Kuhnell D, Benefield D, Haffey W, Wyder M, Patel R, Conrey SC, Burrell AR, Langevin S, Nommsen-Rivers L, Newburg DS, Greis KD, Staat MA, Morrow AL. Prenatal SARS-CoV-2 infection alters postpartum human milk-derived extracellular vesicles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.01.543234. [PMID: 37398231 PMCID: PMC10312504 DOI: 10.1101/2023.06.01.543234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Human milk-derived extracellular vesicles (HMEVs) are crucial functional components in breast milk, contributing to infant health and development. Maternal conditions could affect HMEV cargos; however, the impact of SARS-CoV-2 infection on HMEVs remains unknown. This study evaluated the influence of SARS-CoV-2 infection during pregnancy on postpartum HMEV molecules. Milk samples (9 prenatal SARS-CoV-2 vs. 9 controls) were retrieved from the IMPRINT birth cohort. After defatting and casein micelle disaggregation, 1 mL milk was subjected to a sequential process of centrifugation, ultrafiltration, and qEV-size exclusion chromatography. Particle and protein characterizations were performed following the MISEV2018 guidelines. EV lysates were analyzed through proteomics and miRNA sequencing, while the intact EVs were biotinylated for surfaceomic analysis. Multi-Omics was employed to predict HMEV functions associated with prenatal SARS-CoV-2 infection. Demographic data between the prenatal SARS-CoV-2 and control groups were similar. The median duration from maternal SARS-CoV-2 test positivity to milk collection was 3 months (range: 1-6 months). Transmission electron microscopy showed the cup-shaped nanoparticles. Nanoparticle tracking analysis demonstrated particle diameters of <200 nm and yields of >1e11 particles from 1 mL milk. Western immunoblots detected ALIX, CD9 and HSP70, supporting the presence of HMEVs in the isolates. Thousands of HMEV cargos and hundreds of surface proteins were identified and compared. Multi-Omics predicted that mothers with prenatal SARS-CoV-2 infection produced HMEVs with enhanced functionalities involving metabolic reprogramming and mucosal tissue development, while mitigating inflammation and lower EV transmigration potential. Our findings suggest that SARS-CoV-2 infection during pregnancy boosts mucosal site-specific functions of HMEVs, potentially protecting infants against viral infections. Further prospective studies should be pursued to reevaluate the short- and long-term benefits of breastfeeding in the post-COVID era.
Collapse
|
19
|
Morniroli D, Vizzari G, Tosi M, Treglia G, Corsello A, Marchisio P, Mosca F, Agostoni C, Giannì ML, Milani GP, Pugni L. Mother-to-child transmission of SARS-CoV-2 infection in high-income countries: a systematic review and meta-analysis of prospective observational studies. Sci Rep 2023; 13:8813. [PMID: 37258854 PMCID: PMC10230471 DOI: 10.1038/s41598-023-36097-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 05/29/2023] [Indexed: 06/02/2023] Open
Abstract
Mother-to-child transmission of SARS-CoV-2 has been reported since the onset of the COVID-19 pandemic. We conducted a study to summarize evidence on the risk of mother-to-child transmission in the first 30 days after birth in high-income countries and to evaluate the association between preventive measures and the risk of infection for the neonate. A systematic review and meta-analysis were undertaken following PRISMA guidelines. The National Library of Medicine, Web of Science, and Excerpta Medica databases were screened on February 26, 2022. All prospective observational studies addressing the frequency of infection in infants born to mothers SARS-CoV-2 positive were included. Twenty-six studies were included, reporting data of 2653 mothers with SARS-CoV-2 and 2677 neonates. The proportion meta-analysis pointed out an overall estimate of SARS-CoV-2 infection among infants of 2.3% (95% CI: 1.4-3.2%). Data from studies with (1.4%, 95% CI: 0.8-2) and without (1.3%, 95% CI: 0.0-2.7%) rooming-in provided similar risk of infection. Adopting at least two prevention measures during rooming-in resulted in a rate of mother-to-child infection of 1.0% (95%CI: 0.3-1.7%). The results of this study show a low rate of perinatal infection, support the rooming-in and confirm the effectiveness of preventive measures in reducing the risk of mother-to-child viral transmission.
Collapse
Affiliation(s)
- Daniela Morniroli
- Department of Clinical Sciences and Community Health, University of Milan, via della Commenda 12, 20122, Milan, Italy
| | - Giulia Vizzari
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
| | - Martina Tosi
- Department of Clinical Sciences and Community Health, University of Milan, via della Commenda 12, 20122, Milan, Italy
| | - Giorgio Treglia
- Clinic of Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6500, Bellinzona, Switzerland
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, University of Lausanne, 1015, Lausanne, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900, Lugano, Switzerland
| | - Antonio Corsello
- Department of Clinical Sciences and Community Health, University of Milan, via della Commenda 12, 20122, Milan, Italy
| | - Paola Marchisio
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, S.C. Pediatria-Pneumoinfettivologia, Milan, Lombardia, Italy
- Department of Pathophysiology and Transplantation, University of Milan, 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
| | - Carlo Agostoni
- Department of Clinical Sciences and Community Health, University of Milan, via della Commenda 12, 20122, Milan, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Pediatric Unit, 20122, Milan, Italy
| | - Maria Lorella Giannì
- 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.
| | - Gregorio Paolo Milani
- Department of Clinical Sciences and Community Health, University of Milan, via della Commenda 12, 20122, Milan, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Pediatric Unit, 20122, Milan, Italy
| | - Lorenza Pugni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
| |
Collapse
|
20
|
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
|
21
|
Sajadi MM, Shokatpour N, Purcell M, Tehrani ZR, Lankford A, Bathula A, Campbell JD, Hammershaimb EA, Deatrick KB, Bor C, Parsell DM, Dugan C, Levine AR, Ramelli SC, Chertow DS, Herr DL, Saharia KK, Lewis GK, Grazioli A. Maternal transfer of IgA and IgG SARS-CoV-2 specific antibodies transplacentally and via breast milk feeding. PLoS One 2023; 18:e0284020. [PMID: 37023025 PMCID: PMC10079052 DOI: 10.1371/journal.pone.0284020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 03/21/2023] [Indexed: 04/07/2023] Open
Abstract
BACKGROUND Although there have been many studies on antibody responses to SARS-CoV-2 in breast milk, very few have looked at the fate of these in the infant, and whether they are delivered to immunologically relevant sites in infants. METHODS Mother/infant pairs (mothers who breast milk fed and who were SARS-CoV-2 vaccinated before or after delivery) were recruited for this cross-sectional study. Mother blood, mother breast milk, infant blood, infant nasal specimen, and infant stool was tested for IgA and IgG antibodies against SARS-CoV-2 spike trimer. RESULTS Thirty-one mother/infant pairs were recruited. Breast milk fed infants acquired systemic anti-spike IgG antibodies only if their mothers were vaccinated antepartum (100% Antepartum; 0% Postpartum; P<0.0001). Breast milk fed infants acquired mucosal anti-spike IgG antibodies (in the nose) only if their mothers were vaccinated antepartum (89% Antepartum; 0% Postpartum; P<0.0001). None of the infants in either group had anti-spike IgA in the blood. Surprisingly, 33% of the infants whose mothers were vaccinated antepartum had high titer anti-spike IgA in the nose (33% Antepartum; 0% Postpartum; P = 0.03). Half-life of maternally transferred plasma IgG antibodies in the Antepartum infant cohort was ~70 days. CONCLUSION Vaccination antepartum followed by breast milk feeding appears to be the best way to provide systemic and local anti-SARS-CoV-2 antibodies for infants. The presence of high titer SARS-CoV-2-specific IgA in the nose of infants points to the potential importance of breast milk feeding early in life for maternal transfer of mucosal IgA antibodies. Expectant mothers should consider becoming vaccinated antepartum and consider breast milk feeding for optimal transfer of systemic and mucosal antibodies to their infants.
Collapse
Affiliation(s)
- Mohammad M. Sajadi
- VA Maryland Healthcare Center, Baltimore, MD, United States of America
- Institute of Human Virology, Baltimore, MD, United States of America
| | - Narjes Shokatpour
- Institute of Human Virology, Baltimore, MD, United States of America
| | - Madeleine Purcell
- University of Maryland School of Medicine, Baltimore, MD, United States of America
| | | | - Allison Lankford
- University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Allison Bathula
- University of Maryland Medical Center, Baltimore, MD, United States of America
| | - James D. Campbell
- University of Maryland School of Medicine, Baltimore, MD, United States of America
| | | | | | - Casey Bor
- University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Dawn M. Parsell
- University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Colleen Dugan
- University of Maryland Medical Center, Baltimore, MD, United States of America
| | - Andrea R. Levine
- University of Maryland School of Medicine, Baltimore, MD, United States of America
| | | | - Daniel S. Chertow
- National Institutes of Health, Bethesda, MD, United States of America
| | - Daniel L. Herr
- University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Kapil K. Saharia
- Institute of Human Virology, Baltimore, MD, United States of America
- University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - George K. Lewis
- Institute of Human Virology, Baltimore, MD, United States of America
| | - Alison Grazioli
- University of Maryland School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
22
|
Abstract
The COVID-19 (SARS-Cov-2) pandemic has put a strain on healthcare systems around the world from December 2019 in China, and then rapidly spreading worldwide. The impact of the virus on the entire population and its differential effect on various age groups was unknown at the outset, specifically its severity in elders, children or those living with other comorbidities, thus defining the syndemic, rather than pandemic, character of the infection. The effort of clinicians was initially to organize differential paths to isolate cases or contacts. This impacted the maternal-neonatal care adding an additional burden to this dyad and raising several questions. Can SARS-Cov-2 infection in the first days of life put the health of the newborn at risk? Could the separation of a healthy newborn from an infected mother create further physical and psychological health problems in the dyad? The rapid and massive research effort in these three years of the pandemic has provided wide answers to these initial questions. In this review, we report epidemiological data, clinical features, complications, and management of the neonates affected by SARS-Cov-2 infection.
Collapse
Affiliation(s)
- Alfonso Galderisi
- Department of Woman and Child's Health, University of Padova, Padova, Italy; Yale University, Department of Pediatrics, New Haven, CT, USA
| | - Gianluca Lista
- Neonatal Intensive Care Unit, Buzzi Children's Hospital, Milan, Italy
| | | | - Daniele Trevisanuto
- Department of Woman and Child's Health, University of Padova, Padova, Italy.
| |
Collapse
|
23
|
Chen YY, Tun HM, Field CJ, Mandhane PJ, Moraes TJ, Simons E, Turvey SE, Subbarao P, Scott JA, Kozyrskyj AL. Impact of Cesarean Delivery and Breastfeeding on Secretory Immunoglobulin A in the Infant Gut Is Mediated by Gut Microbiota and Metabolites. Metabolites 2023; 13:metabo13020148. [PMID: 36837767 PMCID: PMC9959734 DOI: 10.3390/metabo13020148] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/19/2022] [Accepted: 01/13/2023] [Indexed: 01/20/2023] Open
Abstract
How gut immunity in early life is shaped by birth in relation to delivery mode, intrapartum antibiotic prophylaxis (IAP) and labor remains undetermined. We aimed to address this gap with a study of secretory Immunoglobulin A (SIgA) in the infant gut that also tested SIgA-stimulating pathways mediated by gut microbiota and metabolites. Among 1017 Canadian full-term infants, gut microbiota of fecal samples collected at 3 and 12 months were profiled using 16S rRNA sequencing; C. difficile was quantified by qPCR; fecal metabolites and SIgA levels were measured by NMR and SIgA enzyme-linked immunosorbent assay, respectively. We assessed the putative causal relationships from birth events to gut microbiota and metabolites, and ultimately to SIgA, in statistical sequential mediation models, adjusted for maternal gravida status in 551 infants. As birth mode influences the ability to breastfeed, the statistical mediating role of breastfeeding status and milk metabolites was also evaluated. Relative to vaginal birth without maternal IAP, cesarean section (CS) after labor was associated with reduced infant gut SIgA levels at 3 months (6.27 vs. 4.85 mg/g feces, p < 0.05); this association was sequentially mediated through gut microbiota and metabolites of microbial or milk origin. Mediating gut microbiota included Enterobacteriaceae, C. difficile, and Streptococcus. The milk or microbial metabolites in CS-SIgA mediating pathways were galactose, fucose, GABA, choline, lactate, pyruvate and 1,2-propanediol. This cohort study documented the impact of birth on infant gut mucosal SIgA. It is the first to characterize gut microbe-metabolite mediated pathways for early-life SIgA maturation, pathways that require experimental verification.
Collapse
Affiliation(s)
- Yuan Yao Chen
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Hein M. Tun
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 1C9, Canada
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Catherine J. Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Piushkumar J. Mandhane
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Theo J. Moraes
- Department of Pediatrics and Physiology, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Elinor Simons
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3A 1S1, Canada
| | - Stuart E. Turvey
- Department of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, BC V6H 0B3, Canada
| | - Padmaja Subbarao
- Department of Pediatrics and Physiology, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - James A. Scott
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Anita L. Kozyrskyj
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 1C9, Canada
- Correspondence: ; Tel.: +1-780-248-5508
| |
Collapse
|
24
|
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: 12] [Impact Index Per Article: 6.0] [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
|
25
|
Garrido-Torres N, Cerrillos L, García Cerro S, Pérez Gómez A, Canal-Rivero M, de Felipe B, Alameda L, Marqués Rodríguez R, Anillo S, Praena J, Duque Sánchez C, Roca C, Paniagua M, López Díaz A, Romero-García R, Olbrich P, Puertas Albarracín MDP, Reguera Pozuelo P, Sosa IL, Moreno Dueñas MB, Pineda Cachero R, Zamudio Juan L, García Rumi V, Guerrero Benitez M, Figueroa R, Martín Rendón AM, Partida A, Rodríguez Cocho MI, Gallardo Trujillo C, Gallego Jiménez I, García Spencer S, Gómez Verdugo M, Bermejo Fernández C, Pérez Benito M, Castillo Reina RE, Cejudo López A, Sánchez Tomás C, Chacón Gamero MÁ, Rubio A, Moreno Mellado A, Ramos Herrero V, Starr E, González Fernández de Palacios M, García Victori E, Pavón Delgado A, Fernández Cuervo I, Arias Ruiz A, Menéndez Gil IE, Domínguez Gómez I, Coca Mendoza I, Ayesa-Arriola R, Fañanas L, Leza JC, Cisneros JM, Sánchez Céspedes J, Ruiz-Mateos E, Crespo-Facorro B, Ruiz-Veguilla M. Examining the immune signatures of SARS-CoV-2 infection in pregnancy and the impact on neurodevelopment: Protocol of the SIGNATURE longitudinal study. Front Pediatr 2022; 10:899445. [PMID: 36619503 PMCID: PMC9811261 DOI: 10.3389/fped.2022.899445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 10/26/2022] [Indexed: 12/24/2022] Open
Abstract
The COVID-19 pandemic represents a valuable opportunity to carry out cohort studies that allow us to advance our knowledge on pathophysiological mechanisms of neuropsychiatric diseases. One of these opportunities is the study of the relationships between inflammation, brain development and an increased risk of suffering neuropsychiatric disorders. Based on the hypothesis that neuroinflammation during early stages of life is associated with neurodevelopmental disorders and confers a greater risk of developing neuropsychiatric disorders, we propose a cohort study of SARS-CoV-2-infected pregnant women and their newborns. The main objective of SIGNATURE project is to explore how the presence of prenatal SARS-CoV-2 infection and other non-infectious stressors generates an abnormal inflammatory activity in the newborn. The cohort of women during the COVID-19 pandemic will be psychological and biological monitored during their pregnancy, delivery, childbirth and postpartum. The biological information of the umbilical cord (foetus blood) and peripheral blood from the mother will be obtained after childbirth. These samples and the clinical characterisation of the cohort of mothers and newborns, are tremendously valuable at this time. This is a protocol report and no analyses have been conducted yet, being currently at, our study is in the recruitment process step. At the time of this publication, we have identified 1,060 SARS-CoV-2 infected mothers and all have already given birth. From the total of identified mothers, we have recruited 537 SARS-COV-2 infected women and all of them have completed the mental health assessment during pregnancy. We have collected biological samples from 119 mothers and babies. Additionally, we have recruited 390 non-infected pregnant women.
Collapse
Affiliation(s)
- Nathalia Garrido-Torres
- Mental Health Unit, Virgen del Rocio University Hospital, Seville, Spain
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
- Spanish Network for Research in Mental Health CIBERSAM, ISCIII, Madrid, Spain
- Department of Psychiatry, University of Seville, Seville, Spain
| | - Lucas Cerrillos
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Susana García Cerro
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
- Spanish Network for Research in Mental Health CIBERSAM, ISCIII, Madrid, Spain
| | - Alberto Pérez Gómez
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Seville Biomedical Research Institute (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, Seville, Spain
| | - Manuel Canal-Rivero
- Mental Health Unit, Virgen del Rocio University Hospital, Seville, Spain
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
- Spanish Network for Research in Mental Health CIBERSAM, ISCIII, Madrid, Spain
- Department of Psychiatry, University of Seville, Seville, Spain
| | - Beatriz de Felipe
- Congenital Immunity Disorders Group de Alteraciones Congénitas de Inmunidad, Seville Biomedical Research Institute, Seville, Spain
- Pediatrics, Infectious Diseases and Immunology Department, University Hospital Virgen del Rocío, Sevilla, Spain
| | - Luis Alameda
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
- Department of Psychiatry, University of Seville, Seville, Spain
- Service of General Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Renata Marqués Rodríguez
- Department of Pediatrics, Virgen del Rocío University Hospital / Institute of Biomedicine of Seville (IBiS), Seville, Spain
| | - Sergio Anillo
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Julia Praena
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Cristina Duque Sánchez
- Department of Pediatrics, Virgen del Rocío University Hospital / Institute of Biomedicine of Seville (IBiS), Seville, Spain
| | - Cristina Roca
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Seville Biomedical Research Institute (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, Seville, Spain
| | - María Paniagua
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Seville Biomedical Research Institute (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, Seville, Spain
| | - Alvaro López Díaz
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
- Spanish Network for Research in Mental Health CIBERSAM, ISCIII, Madrid, Spain
- Department of Psychiatry, University of Seville, Seville, Spain
| | - Rafael Romero-García
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
- Spanish Network for Research in Mental Health CIBERSAM, ISCIII, Madrid, Spain
- Department of Medical Physiology and Biophysics, Seville Biomedical Research Institute (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, Seville, Spain
| | - Peter Olbrich
- Congenital Immunity Disorders Group de Alteraciones Congénitas de Inmunidad, Seville Biomedical Research Institute, Seville, Spain
| | | | - Pablo Reguera Pozuelo
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
| | - Irene Luján Sosa
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - María Begoña Moreno Dueñas
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Rocío Pineda Cachero
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Lidia Zamudio Juan
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Verónica García Rumi
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Mercedes Guerrero Benitez
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Rosario Figueroa
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Antonio Manuel Martín Rendón
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Antonio Partida
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - María Isabel Rodríguez Cocho
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Carmen Gallardo Trujillo
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Isabel Gallego Jiménez
- Department of Genetics, Reproduction and Maternal-Fetal Medicine, University Hospital Virgen del Rocío, Seville, Spain
| | - Sarah García Spencer
- Department of Pediatrics, Virgen del Rocío University Hospital / Institute of Biomedicine of Seville (IBiS), Seville, Spain
| | - Marta Gómez Verdugo
- Department of Pediatrics, Virgen del Rocío University Hospital / Institute of Biomedicine of Seville (IBiS), Seville, Spain
| | - Cintia Bermejo Fernández
- Department of Pediatrics, Virgen del Rocío University Hospital / Institute of Biomedicine of Seville (IBiS), Seville, Spain
| | - María Pérez Benito
- Department of Pediatrics, Virgen del Rocío University Hospital / Institute of Biomedicine of Seville (IBiS), Seville, Spain
| | | | - Angela Cejudo López
- Department of family medicine, Virgen del Rocío University Hospital, Primary Care Health Centers, Seville, Spain
| | - Candela Sánchez Tomás
- Department of family medicine, Virgen del Rocío University Hospital, Primary Care Health Centers, Seville, Spain
| | | | - Ana Rubio
- Mental Health Unit, Virgen del Rocio University Hospital, Seville, Spain
| | - Amanda Moreno Mellado
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
| | - Víctor Ramos Herrero
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
| | - Ella Starr
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
| | | | - Elena García Victori
- Department of Pediatrics, Virgen del Rocío University Hospital / Institute of Biomedicine of Seville (IBiS), Seville, Spain
| | - Antonio Pavón Delgado
- Department of Pediatrics, Virgen del Rocío University Hospital / Institute of Biomedicine of Seville (IBiS), Seville, Spain
| | | | | | | | | | | | - Rosa Ayesa-Arriola
- Spanish Network for Research in Mental Health CIBERSAM, ISCIII, Madrid, Spain
- Department of Psychiatry, University Hospital Marqués de Valdecilla - Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Lourdes Fañanas
- Spanish Network for Research in Mental Health CIBERSAM, ISCIII, Madrid, Spain
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology, University of Barcelona (UB), Barcelona, Spain
| | - Juan C Leza
- Spanish Network for Research in Mental Health CIBERSAM, ISCIII, Madrid, Spain
- Department of Pharmacology & Toxicology, Faculty of Medicine, Universidad Complutense Madrid, CIBERSAM, Imas12, IUINQ, Madrid, Spain
| | - José M Cisneros
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Seville Biomedical Research Institute (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, Seville, Spain
| | - Javier Sánchez Céspedes
- Viral Diseases and Infections in Immunodeficiencies Research Group, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Seville, Spain
| | - Ezequiel Ruiz-Mateos
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Seville Biomedical Research Institute (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, Seville, Spain
| | - Benedicto Crespo-Facorro
- Mental Health Unit, Virgen del Rocio University Hospital, Seville, Spain
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
- Spanish Network for Research in Mental Health CIBERSAM, ISCIII, Madrid, Spain
- Department of Psychiatry, University of Seville, Seville, Spain
| | - Miguel Ruiz-Veguilla
- Mental Health Unit, Virgen del Rocio University Hospital, Seville, Spain
- Translational Psychiatry Group, Seville Biomedical Research Institute (IBiS), Seville, Spain
- Spanish Network for Research in Mental Health CIBERSAM, ISCIII, Madrid, Spain
- Department of Psychiatry, University of Seville, Seville, Spain
| |
Collapse
|
26
|
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
|
27
|
Valenzuela-Fernández A, Cabrera-Rodriguez R, Ciuffreda L, Perez-Yanes S, Estevez-Herrera J, González-Montelongo R, Alcoba-Florez J, Trujillo-González R, García-Martínez de Artola D, Gil-Campesino H, Díez-Gil O, Lorenzo-Salazar JM, Flores C, Garcia-Luis J. Nanomaterials to combat SARS-CoV-2: Strategies to prevent, diagnose and treat COVID-19. Front Bioeng Biotechnol 2022; 10:1052436. [PMID: 36507266 PMCID: PMC9732709 DOI: 10.3389/fbioe.2022.1052436] [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: 09/23/2022] [Accepted: 11/09/2022] [Indexed: 11/26/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the associated coronavirus disease 2019 (COVID-19), which severely affect the respiratory system and several organs and tissues, and may lead to death, have shown how science can respond when challenged by a global emergency, offering as a response a myriad of rapid technological developments. Development of vaccines at lightning speed is one of them. SARS-CoV-2 outbreaks have stressed healthcare systems, questioning patients care by using standard non-adapted therapies and diagnostic tools. In this scenario, nanotechnology has offered new tools, techniques and opportunities for prevention, for rapid, accurate and sensitive diagnosis and treatment of COVID-19. In this review, we focus on the nanotechnological applications and nano-based materials (i.e., personal protective equipment) to combat SARS-CoV-2 transmission, infection, organ damage and for the development of new tools for virosurveillance, diagnose and immune protection by mRNA and other nano-based vaccines. All the nano-based developed tools have allowed a historical, unprecedented, real time epidemiological surveillance and diagnosis of SARS-CoV-2 infection, at community and international levels. The nano-based technology has help to predict and detect how this Sarbecovirus is mutating and the severity of the associated COVID-19 disease, thereby assisting the administration and public health services to make decisions and measures for preparedness against the emerging variants of SARS-CoV-2 and severe or lethal COVID-19.
Collapse
Affiliation(s)
- Agustín Valenzuela-Fernández
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Romina Cabrera-Rodriguez
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Laura Ciuffreda
- Research Unit, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Silvia Perez-Yanes
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Judith Estevez-Herrera
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | | | - Julia Alcoba-Florez
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Rodrigo Trujillo-González
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
- Departamento de Análisis Matemático, Facultad de Ciencias, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | | | - Helena Gil-Campesino
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Oscar Díez-Gil
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - José M. Lorenzo-Salazar
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
| | - Carlos Flores
- Research Unit, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Faculty of Health Sciences, University of Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| | - Jonay Garcia-Luis
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| |
Collapse
|
28
|
Piñeiro-Salvador R, Vazquez-Garza E, Cruz-Cardenas JA, Licona-Cassani C, García-Rivas G, Moreno-Vásquez J, Alcorta-García MR, Lara-Diaz VJ, Brunck MEG. A cross-sectional study evidences regulations of leukocytes in the colostrum of mothers with obesity. BMC Med 2022; 20:388. [PMID: 36316769 PMCID: PMC9624055 DOI: 10.1186/s12916-022-02575-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Breastmilk is a dynamic fluid whose initial function is to provide the most adapted nutrition to the neonate. Additional attributes have been recently ascribed to breastmilk, with the evidence of a specific microbiota and the presence of various components of the immune system, such as cytokines and leukocytes. The composition of breastmilk varies through time, according to the health status of mother and child, and altogether contributes to the future health of the infant. Obesity is a rising condition worldwide that creates a state of systemic, chronic inflammation including leukocytosis. Here, we asked whether colostrum, the milk produced within the first 48 h post-partum, would contain a distinct leukocyte composition depending on the body mass index (BMI) of the mother. METHODS We collected peripheral blood and colostrum paired samples from obese (BMI > 30) and lean (BMI < 25) mothers within 48 h post-partum and applied a panel of 6 antibodies plus a viability marker to characterize 10 major leukocyte subpopulations using flow cytometry. RESULTS The size, internal complexity, and surface expression of CD45 and CD16 of multiple leukocyte subpopulations were selectively regulated between blood and colostrum irrespective of the study groups, suggesting a generalized cell-specific phenotype alteration. In obesity, the colostrum B lymphocyte compartment was significantly reduced, and CD16+ blood monocytes had an increased CD16 expression compared to the lean group. CONCLUSIONS This is the first characterization of major leukocyte subsets in colostrum of mothers suffering from obesity and the first report of colostrum leukocyte subpopulations in Latin America. We evidence various significant alterations of most leukocyte populations between blood and colostrum and demonstrate a decreased colostrum B lymphocyte fraction in obesity. This pioneering study is a stepping stone to further investigate active immunity in human breastmilk.
Collapse
Affiliation(s)
- Raúl Piñeiro-Salvador
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Av. Eugenio Garza Sada 2501 Sur, Tecnologico, 64849, Monterrey, Nuevo León, Mexico
| | - Eduardo Vazquez-Garza
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000 Poniente, Col. Doctores, 64710, Monterrey, Nuevo León, Mexico
| | - José Antonio Cruz-Cardenas
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Av. Eugenio Garza Sada 2501 Sur, Tecnologico, 64849, Monterrey, Nuevo León, Mexico
| | - Cuauhtémoc Licona-Cassani
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Av. Eugenio Garza Sada 2501 Sur, Tecnologico, 64849, Monterrey, Nuevo León, Mexico
- The Institute for Obesity Research, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Tecnologico, 64849, Monterrey, Nuevo León, Mexico
| | - Gerardo García-Rivas
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000 Poniente, Col. Doctores, 64710, Monterrey, Nuevo León, Mexico
- The Institute for Obesity Research, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Tecnologico, 64849, Monterrey, Nuevo León, Mexico
| | - Jorge Moreno-Vásquez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000 Poniente, Col. Doctores, 64710, Monterrey, Nuevo León, Mexico
| | - Mario René Alcorta-García
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000 Poniente, Col. Doctores, 64710, Monterrey, Nuevo León, Mexico
- Hospital Regional Materno-Infantil, SSNL, OPD, Ciudad Guadalupe, Nuevo León, Mexico
| | - Victor Javier Lara-Diaz
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000 Poniente, Col. Doctores, 64710, Monterrey, Nuevo León, Mexico
| | - Marion E G Brunck
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Av. Eugenio Garza Sada 2501 Sur, Tecnologico, 64849, Monterrey, Nuevo León, Mexico.
- The Institute for Obesity Research, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Tecnologico, 64849, Monterrey, Nuevo León, Mexico.
| |
Collapse
|
29
|
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
|
30
|
Calabretto M, D'Alisa R, Faraone S, Mazzuti L, Pecorini F, Turriziani O. Detection of SARS-CoV-2 RNA and antibodies in breast milk of infected mothers. J Med Virol 2022; 95:e28142. [PMID: 36098372 PMCID: PMC9538366 DOI: 10.1002/jmv.28142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/19/2022] [Accepted: 09/09/2022] [Indexed: 01/11/2023]
Affiliation(s)
| | - Rossella D'Alisa
- Department of Maternal Infantile and Urological SciencesSapienza University of RomeRomeItaly
| | - Sara Faraone
- Department of Molecular MedicineSapienza University of RomeRomeItaly
| | - Laura Mazzuti
- Department of Molecular MedicineSapienza University of RomeRomeItaly
| | - Francesco Pecorini
- Department of Maternal Infantile and Urological SciencesSapienza University of RomeRomeItaly
| | | |
Collapse
|
31
|
Rostomian L, Angelidou A, Sullivan K, Melvin PR, Shui JE, Telefus Goldfarb I, Bartolome R, Chaudhary N, Singh R, Vaidya R, Steele T, Yanni D, Patrizi S, Culic I, Parker MG, Belfort MB. The Effects of COVID-19 Hospital Practices on Breastfeeding Initiation and Duration Postdischarge. Breastfeed Med 2022; 17:736-744. [PMID: 35731120 DOI: 10.1089/bfm.2022.0039] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background: Early in the COVID-19 pandemic, many birth hospitals separated SARS-CoV-2-positive mothers from their newborn infants and advised against breastfeeding to decrease postnatal SARS-CoV-2 transmission. Information on how these practices impacted breastfeeding postdischarge is limited. Objectives: In a statewide sample of SARS-CoV-2-positive mothers, we aimed to determine the extent to which (1) mother-infant separation and (2) a lack of breastfeeding initiation in-hospital were associated with breast milk feeding postdischarge. Design/Methods: From 11 birthing hospitals in Massachusetts, we identified 187 women who tested positive for SARS-CoV-2 from 14 days before to 72 hours after delivery (March 1-July 31, 2020) and their newborn infants. We abstracted chart data from the delivery hospitalization on main exposure variables (mother-infant separation, in-hospital breast milk feeding [expressed milk feeding and/or direct breastfeeding]) and from outpatient visits until 30 days postdischarge. We evaluated associations of in-hospital practices with outcomes up to 30 days postdischarge, adjusting for confounders using multivariable logistic and linear regression. Results: Mother-infant separation in-hospital was associated with a shorter duration of any breast milk feeding (regression coefficient estimate -5.29 days, 95% confidence intervals [CI] [-8.89 to -1.69]). Direct breastfeeding in-hospital was associated with higher odds of any breast milk feeding (adjusted odds ratios [AOR] 5.68, 95% CI [1.65-23.63]) and direct breastfeeding (AOR 8.19, 95% CI [2.99-24.91]) postdischarge; results were similar for any breast milk feeding in-hospital. Conclusions: Perinatal hospital care practices implemented early in the COVID-19 pandemic, specifically mother-infant separation and prevention of breast milk feeding initiation, were associated with adverse effects on breast milk feeding outcomes assessed up to 1 month postdischarge.
Collapse
Affiliation(s)
- Lara Rostomian
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Asimenia Angelidou
- Department of Neonatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Katherine Sullivan
- UMass Memorial Health Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Patrice R Melvin
- Office of Health Equity and Inclusion, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Jessica E Shui
- Division of Newborn Medicine, Department of Pediatrics, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ilona Telefus Goldfarb
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts
| | - Ruby Bartolome
- Department of Pediatrics, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Neha Chaudhary
- Division of Newborn Medicine, Tufts Children's Hospital, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Rachana Singh
- Division of Newborn Medicine, Tufts Children's Hospital, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Ruben Vaidya
- Department of Pediatrics, UMass Chan Medical School-Baystate, Springfield, Massachusetts, USA
| | - Tina Steele
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Diana Yanni
- Department of Neonatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Silvia Patrizi
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Ivana Culic
- Department of Neonatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Margaret G Parker
- Department of Pediatrics, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Mandy B Belfort
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts. USA
| |
Collapse
|
32
|
Gandecha H, Kaur A, Sanghera R, Preece J, Pillay T. Nutrition and Immunity in Perinatal Hypoxic-Ischemic Injury. Nutrients 2022; 14:nu14132747. [PMID: 35807927 PMCID: PMC9269416 DOI: 10.3390/nu14132747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/23/2022] [Accepted: 06/28/2022] [Indexed: 02/01/2023] Open
Abstract
Perinatal hypoxia ischaemia (PHI), acute and chronic, may be associated with considerable adverse outcomes in the foetus and neonate. The molecular and cellular mechanisms of injury and repair associated with PHI in the perinate are not completely understood. Increasing evidence is mounting for the role of nutrients and bioactive food components in immune development, function and repair in PHI. In this review, we explore current concepts around the neonatal immune response to PHI with a specific emphasis on the impact of nutrition in the mother, foetus and neonate.
Collapse
Affiliation(s)
- Hema Gandecha
- Department of Neonatology, University Hospitals Leicester NHS Trust, Leicester LE1 5WW, UK
- East Midlands Deanery, Health Education England, Leicester LE3 5DR, UK
| | - Avineet Kaur
- Department of Neonatology, University Hospitals Leicester NHS Trust, Leicester LE1 5WW, UK
- East Midlands Deanery, Health Education England, Leicester LE3 5DR, UK
| | - Ranveer Sanghera
- Department of Neonatology, University Hospitals Leicester NHS Trust, Leicester LE1 5WW, UK
- East Midlands Deanery, Health Education England, Leicester LE3 5DR, UK
| | - Joanna Preece
- Department of Neonatology, University Hospitals Leicester NHS Trust, Leicester LE1 5WW, UK
| | - Thillagavathie Pillay
- Department of Neonatology, University Hospitals Leicester NHS Trust, Leicester LE1 5WW, UK
- Faculty of Science and Engineering, Research Institute for Healthcare Sciences, University of Wolverhampton, Wolverhampton WV1 1LY, UK
- College of Life Sciences, University of Leicester, Leicester LE5 4PW, UK
| |
Collapse
|
33
|
Humoral response to anti-SARS-CoV-2 vaccine in breastfeeding mothers and mother-to-infant antibody transfer through breast milk. NPJ Vaccines 2022; 7:63. [PMID: 35739127 PMCID: PMC9226003 DOI: 10.1038/s41541-022-00499-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/16/2022] [Indexed: 11/08/2022] Open
Abstract
The magnitude of mother-to-infant transfer of anti-SARS-CoV-2 antibodies through breast milk (BM) after maternal vaccination during breastfeeding, in the absence of transplacental transfer of IgG, remains unclear. Here, we quantified anti-S and anti-RBD IgG, IgA, IgA1, and IgA2 in maternal serum, maternal saliva, BM, infant buccal swabs, and infant feces up to 90 days after the second maternal vaccine dose. BNT162b2 vaccine induced long-lasting IgG in maternal serum, but weaker mucosal antibody production, with anti-SARS-CoV-2 IgG and IgA amounts in BM between 10- and 150-fold lower compared to serum. BM IgA were exclusively of the IgA1 isotype, with no production of the mucosal-specific and protease-resistant IgA2. Accordingly, only traces of antibodies were retrieved from the feces of breastfed infants, and no IgG nor IgA were retrieved from infants' buccal swabs. Newly engineered anti-SARS-CoV-2 vaccines may be needed to stimulate the antibody production at mucosal sites such as breast milk.
Collapse
|
34
|
Pang Z, Hu R, Tian L, Lou F, Chen Y, Wang S, He S, Zhu S, An X, Song L, Liu F, Tong Y, Fan H. Overview of Breastfeeding Under COVID-19 Pandemic. Front Immunol 2022; 13:896068. [PMID: 35711421 PMCID: PMC9192965 DOI: 10.3389/fimmu.2022.896068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/22/2022] [Indexed: 12/19/2022] Open
Abstract
During the global pandemic of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), pregnant and lactating women are at higher risk of infection. The potential of viral intrauterine transmission and vertical transmission by breastfeeding has raised wide concerns. Breastmilk is rich in nutrients that contribute to infant growth and development, and reduce the incidence rate of infant illness and death, as well as inhibit pathogens significantly, and protect infants from infection. Although it is controversial whether mothers infected with COVID-19 should continue to breastfeed, many countries and international organizations have provided recommendations and guidance for breastfeeding. This review presents the risks and benefits of breastfeeding for mothers infected with COVID-19, and the reasons for the absence of SARS-CoV-2 active virus in human milk. In addition, the antiviral mechanisms of nutrients in breastmilk, the levels of SARS-CoV-2 specific antibodies in breastmilk from COVID-19 infected mothers and vaccinated mothers are also summarized and discussed, aiming to provide some support and recommendations for both lactating mothers and infants to better deal with the COVID-19 pandemic.
Collapse
Affiliation(s)
- Zehan Pang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Ruolan Hu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Lili Tian
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Fuxing Lou
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Yangzhen Chen
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Shuqi Wang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Shiting He
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Shaozhou Zhu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Xiaoping An
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Lihua Song
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Feitong Liu
- Health & Happiness Group, Health & Happiness Research, China Aesearch and Innovation, Guangzhou, China
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Huahao Fan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| |
Collapse
|
35
|
Abstract
Extremely preterm infants are particularly vulnerable to systemic infections secondary to their immature immune defenses, prolonged hospitalizations, delays in enteral feeding, early antibiotic exposure, and need for life-sustaining invasive interventions. There have been several evidence-based practices for infection prevention in this population, such as human milk feedings, utilization of "bundle checklists" and decolonization of pathogenic organisms. Other practices, such as the use of probiotics, human milk-derived fortifiers, and antifungal prophylaxis are more controversial and require further investigation regarding the risks and benefits of such interventions. This chapter examines the susceptibility of the preterm newborn infant to invasive infections and describes several strategies for infection prevention, along with the associated limitations of such practices. It also addresses the various gaps in our understanding of preventing infections in this population, and the need for additional large multi-center randomized controlled trials. Additionally, the role of the SARs-CoV-2 global pandemic and associated strategies for infection prevention in the NICU are discussed.
Collapse
|
36
|
Conti MG, Terreri S, Terrin G, Natale F, Pietrasanta C, Salvatori G, Brunelli R, Midulla F, Papaevangelou V, Carsetti R, Angelidou A. Severe Acute Respiratory Syndrome Coronavirus 2 Infection Versus Vaccination in Pregnancy: Implications for Maternal and Infant Immunity. Clin Infect Dis 2022; 75:S37-S45. [PMID: 35535796 PMCID: PMC9129222 DOI: 10.1093/cid/ciac359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/22/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been associated with adverse maternal and neonatal outcomes, yet uptake of SARS-CoV-2 vaccines during pregnancy and lactation has been slow. As a result, millions of pregnant and lactating women and their infants remain susceptible to the virus. METHODS We measured spike-specific immunoglobulin G (anti-S IgG) and immunoglobulin A (anti-S IgA) in serum and breastmilk (BM) samples from 3 prospective mother-infant cohorts recruited in 2 academic medical centers. The primary aim was to determine the impact of maternal SARS-CoV-2 immunization vs infection and their timing on systemic and mucosal immunity. RESULTS The study included 28 mothers infected with SARS-CoV-2 in late pregnancy (INF), 11 uninfected mothers who received 2 doses of the BNT162b2 vaccine in the latter half of pregnancy (VAX-P), and 12 uninfected mothers who received 2 doses of BNT162b2 during lactation. VAX dyads had significantly higher serum anti-S IgG compared to INF dyads (P < .0001), whereas INF mothers had higher BM:serum anti-S IgA ratios compared to VAX mothers (P = .0001). Median IgG placental transfer ratios were significantly higher in VAX-P compared to INF mothers (P < .0001). There was a significant positive correlation between maternal and neonatal serum anti-S IgG after vaccination (r = 0.68, P = .013), but not infection. CONCLUSIONS BNT161b2 vaccination in late pregnancy or lactation enhances systemic immunity through serum anti-S immunoglobulin, while SARS-CoV-2 infection induces mucosal over systemic immunity more efficiently through BM immunoglobulin production. Next-generation vaccines boosting mucosal immunity could provide additional protection to the mother-infant dyad. Future studies should focus on identifying the optimal timing of primary and/or booster maternal vaccination for maximal benefit.
Collapse
Affiliation(s)
- Maria Giulia Conti
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Sara Terreri
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Gianluca Terrin
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Fabio Natale
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Carlo Pietrasanta
- NICU, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Guglielmo Salvatori
- Neonatal Intensive Care Unit and Human Milk Bank, Department of Neonatology, Bambino Gesù Children’s Hospital, IRCSS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy
| | - Roberto Brunelli
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Fabio Midulla
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Vassiliki Papaevangelou
- Third Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| | - Rita Carsetti
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy,Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children's Hospital, IRCCS; Piazza Sant’Onofrio, 4, 00165, Rome, Italy
| | - Asimenia Angelidou
- Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, United States,Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital,Harvard Medical School,Corresponding author: Asimenia Angelidou, MD PhD Instructor in Pediatrics, Harvard Medical School 4 Blackfan Circle, HIM Building, Rm 836, Boston MA 02115
| |
Collapse
|
37
|
Enengl S, Pecks U, Oppelt P, Stelzl P, Trautner PS, Shebl O, Lamprecht B, Longardt AC, Eckmann-Scholz C, Keil C, Mand N, von Kaisenberg CS, Jegen M, Doppler S, Lastinger J. Antibody Response and Maternofetal Antibody Transfer in SARS-CoV-2-Positive Pregnant Women: A Multicenter Observational Study. Geburtshilfe Frauenheilkd 2022; 82:501-509. [PMID: 35528188 PMCID: PMC9076216 DOI: 10.1055/a-1768-0415] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 02/08/2022] [Indexed: 12/20/2022] Open
Abstract
Introduction Awareness of SARS-CoV-2 infection in pregnant women and the potential risk for infection of their neonates is increasing. The aim of this study was to examine the immune status of affected women and evaluate the dynamics of placental antibody transfer. Materials and Methods The study included 176 women with SARS-CoV-2 infection during pregnancy who delivered between April 2020 and December 2021 at eight obstetric maternity sites. Demographic data, maternal and neonatal characteristics were summarized. Antibody testing for IgA and IgG in maternal blood sera and umbilical cord samples was evaluated and IgG transfer ratios were calculated. Values were related to the time of infection during pregnancy and birth. Results The percentage of IgG positive women increased from 29.0% (95% CI 23.8 - 37.8) at presentation with a positive PCR test result to 75.7% (95% CI 71.6 - 79.8), the percentage of IgG positive umbilical cord blood samples increased from 17.1% (95% CI 13.0 - 21.3) to 76.4% (95% CI 72.2 - 80.7) at more than six weeks after infection. Regression lines differed significantly between maternal and fetal IgG responses (p < 0.0001). Newborns react with a latency of about one week; umbilical cord blood antibody concentrations are highly correlated with maternal concentration levels (ρ = 0.8042; p < 0.0001). IgG transplacental transfer ratios were dependent on infection-to-birth interval. Two of the umbilical cord blood samples tested positive for IgA. Conclusions These findings confirm vertical SARS-CoV-2 transmission is rare; however, antibodies are transferred to the fetus soon after infection during pregnancy. Since transplacental antibody transfer might have a protective value for neonatal immunization this information may be helpful when counseling affected women.
Collapse
Affiliation(s)
- Sabine Enengl
- Department of Gynecology, Obstetrics, and Gynecological Endocrinology, Kepler University Hospital, Johannes Kepler University Linz, Linz, Austria
| | - Ulrich Pecks
- Department of Obstetrics, Schleswig-Holstein University Hospital, Kiel, Germany
| | - Peter Oppelt
- Department of Gynecology, Obstetrics, and Gynecological Endocrinology, Kepler University Hospital, Johannes Kepler University Linz, Linz, Austria
| | - Patrick Stelzl
- Department of Gynecology, Obstetrics, and Gynecological Endocrinology, Kepler University Hospital, Johannes Kepler University Linz, Linz, Austria
| | - Philip Sebastian Trautner
- Department of Gynecology, Obstetrics, and Gynecological Endocrinology, Kepler University Hospital, Johannes Kepler University Linz, Linz, Austria
| | - Omar Shebl
- Department of Gynecology, Obstetrics, and Gynecological Endocrinology, Kepler University Hospital, Johannes Kepler University Linz, Linz, Austria
| | - Bernd Lamprecht
- Department of Pulmonology, Kepler University Hospital, Johannes Kepler University Linz, Linz, Austria
| | | | | | - Corinna Keil
- Department of Obstetrics, Philipps University of Marburg, Marburg, Germany
| | - Nadine Mand
- Department of Pediatrics, Philipps University of Marburg, Marburg, Germany
| | | | - Magdalena Jegen
- Department of Obstetrics and Gynecology, University Hospital, LMU, Munich, Germany
| | - Stefan Doppler
- Department of Pathology, Kepler University Hospital, Johannes Kepler University Linz, Linz, Austria
| | - Julia Lastinger
- Department of Gynecology, Obstetrics, and Gynecological Endocrinology, Kepler University Hospital, Johannes Kepler University Linz, Linz, Austria
| |
Collapse
|
38
|
G.R. Q. The Impact of the SARS-CoV-2 Pandemic on Reproduction, Sexual Function and Behaviors: A Review of the Main Trends and Findings. INTERNATIONAL JOURNAL OF SEXUAL HEALTH : OFFICIAL JOURNAL OF THE WORLD ASSOCIATION FOR SEXUAL HEALTH 2022; 34:351-365. [PMID: 38596275 PMCID: PMC10903693 DOI: 10.1080/19317611.2022.2053921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 01/31/2022] [Accepted: 03/07/2022] [Indexed: 04/11/2024]
Abstract
Objective: This review sought to describe the main trends and findings on the SARS-CoV-2 and the pandemic's effect on reproduction (i.e., effects on fertility, reproductive tissue, pregnancy, vertical and sexual transmission) and sexual function and behaviors. Methods: A review was conducted on studies assessing these variables through the Scopus and PubMed databases between the years 2019-2021. Results: Results showed SARS-CoV-2 pandemic and social distancing measures have affected-and in some cases not-different aspects of people's reproductive function, sexual function and behaviors. Conclusions: Suggestions are offered for researchers and professionals focused on future research and clinical practice.
Collapse
Affiliation(s)
- Quintana G.R.
- Departamento de Psicología y Filosofía, Facultad de Ciencias Sociales y Jurídicas, Universidad de Tarapacá, Arica, Arica y Parinacota, Chile
| |
Collapse
|
39
|
Long-term infant feeding roles in triggering uncontrolled inflammatory responses. Pediatr Res 2022; 92:340. [PMID: 35654834 PMCID: PMC9179222 DOI: 10.1038/s41390-022-02131-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 11/09/2022]
|
40
|
Microbial antigen in human milk: a natural vaccine? Mucosal Immunol 2022; 15:1058-1059. [PMID: 36030351 PMCID: PMC9419140 DOI: 10.1038/s41385-022-00561-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/26/2022] [Accepted: 08/10/2022] [Indexed: 02/04/2023]
|