1
|
Mane V, Mehta R, Alvarez N, Sharma V, Park S, Fox A, DeCarlo C, Yang X, Perlin DS, Powell RLR. In vivo antiviral efficacy of LCTG-002, a pooled, purified human milk secretory IgA product, against SARS-CoV-2 in a murine model of COVID-19. Hum Vaccin Immunother 2024; 20:2303226. [PMID: 38251677 PMCID: PMC10807469 DOI: 10.1080/21645515.2024.2303226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
Immunoglobulin A (IgA) is the most abundant antibody (Ab) in human mucosae, with secretory form (sIgA) being dominant and uniquely stable. sIgA is challenging to produce recombinantly but is naturally found in human milk, which could be considered a global resource for this biologic, justifying its development as a mucosal therapeutic. Presently, SARS-CoV-2 was utilized as a model mucosal pathogen, and methods were developed to efficiently extract human milk sIgA from donors who were naïve to SARS-CoV-2 or had recovered from infection that elicited high-titer anti-SARS-CoV-2 Spike sIgA in their milk (pooled to make LCTG-002). Mass spectrometry determined that proteins with a relative abundance of 1% or greater were all associated with sIgA. Western blot demonstrated that all batches consisted predominantly of sIgA. Compared to control IgA, LCTG-002 demonstrated significantly higher Spike binding (mean endpoint of 0.87 versus 5.87). LCTG-002 was capable of blocking the Spike receptor-binding domain - angiotensin-converting enzyme 2 (ACE2) interaction with significantly greater potency compared to control (mean LCTG-002 IC50 154ug/mL versus 50% inhibition not achieved for control), and exhibited significant neutralization activity against Spike-pseudotyped virus infection (mean LCTG-002 IC50 49.8ug/mL versus 114.5ug/mL for control). LCTG-002 was tested for its capacity to reduce viral lung burden in K18+hACE2 transgenic mice inoculated with SARS-CoV-2. LCTG-002 significantly reduced SARS-CoV-2 titers compared to control when administered at 0.25 mg/day or 1 mg/day, with a maximum TCID50 reduction of 4.9 logs. This innovative study demonstrates that LCTG-002 is highly pure and efficacious in vivo, supporting further development of milk-derived, polyclonal sIgA therapeutics.
Collapse
Affiliation(s)
- Viraj Mane
- Lactiga US, Inc. 675 US-1, North Brunswick, NJ, USA
| | - Rikin Mehta
- Lactiga US, Inc. 675 US-1, North Brunswick, NJ, USA
| | - Nadine Alvarez
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Vijeta Sharma
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Steven Park
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Alisa Fox
- Icahn School of Medicine at Mount Sinai, Department of Medicine, Division of Infectious Diseases, New York, NY, USA
| | - Claire DeCarlo
- Icahn School of Medicine at Mount Sinai, Department of Medicine, Division of Infectious Diseases, New York, NY, USA
| | - Xiaoqi Yang
- Icahn School of Medicine at Mount Sinai, Department of Medicine, Division of Infectious Diseases, New York, NY, USA
| | - David S. Perlin
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Rebecca L. R. Powell
- Icahn School of Medicine at Mount Sinai, Department of Medicine, Division of Infectious Diseases, New York, NY, USA
| |
Collapse
|
2
|
Korchinski I, Marquez C, McClymont E, Av-Gay G, Andrade J, Elwood C, Jassem A, Krajden M, Morshed M, Sadarangani M, Tanunliong G, Sekirov I, Money D. Maternal-infant transfer of SARS-CoV-2 antibodies following vaccination in pregnancy: A prospective cohort study. Vaccine 2024; 42:126123. [PMID: 38981741 DOI: 10.1016/j.vaccine.2024.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/29/2024] [Accepted: 07/05/2024] [Indexed: 07/11/2024]
Abstract
OBJECTIVES To measure and evaluate the impact of receiving severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines in pregnancy on immunoglobulin G (IgG) and immunoglobulin A (IgA) titres in maternal and infant samples. DESIGN Prospective cohort study. SETTING Tertiary obstetric centre. POPULATION OR SAMPLE 52 pregnant women who received one or more SARS-CoV-2 vaccine doses during pregnancy and their neonates. METHODS IgG and IgA concentrations against SARS-CoV-2 antigens were measured from samples collected at delivery and 4-6 weeks postpartum and compared using Spearman correlations. MAIN OUTCOME MEASURES Maternal and infant IgG and IgA titres in response to vaccination and infection in pregnancy. RESULTS In maternal serum collected at delivery, participants without evidence of prior infection who received 3 + doses of a SARS-CoV-2 vaccine had higher Anti-Spike (S) IgG geometric mean concentrations (log10 AU/mL)(GMC) than those who received 2 doses (3 + Doses = 5.00, 2 Doses = 4.60, p = 0.08). The differences in IgG Anti-S GMC were statistically significant in cord serum, and in postpartum samples of maternal serum, infant serum and breast milk (Cord GMCs: 3 + Doses = 5.32, 2 Doses = 4.98, p < 0.05; Postpartum maternal serum GMCs: 3 + Doses = 5.25, 2 Doses = 4.57, p < 0.001; Postpartum infant serum GMCs: 3 + Doses = 5.10, 2 Doses = 4.72, p = 0.03; Postpartum breast milk GMCs: 3 + Doses = 2.61, 2 Doses = 1.94, p < 0.0001). Among participants with 3 + Doses, those with evidence of SARS-CoV-2 infection had statistically significant higher anti-S IgG GMCs than those without prior infection (Maternal serum at delivery: SARS-CoV-2+=5.65, SARS-CoV-2-=5.00, p = 0.004; Cord: SARS-CoV-2+=5.68, SARS-CoV-2-=5.32, p = 0.02; Postpartum maternal serum: SARS-CoV-2+=5.66, SARS-CoV-2-=5.25, p < 0.001; postpartum infant serum: SARS-CoV-2+=5.50, SARS-CoV-2-=5.10, p = 0.003; Postpartum breast milk: SARS-COV-2+=3.25, SARS-COV-2-=2.61, p = 0.009). Significant positive correlations were found for anti-S IgG titres between paired maternal and infant samples at delivery and postpartum (Delivery: R = 0.91, p < 0.001; postpartum: R = 0.86, p < 0.001). CONCLUSIONS Receipt of a SARS-CoV-2 vaccine and SARS-CoV-2 infection elicit strong IgG and IgA antibody responses in pregnant women with evidence of transplacental transfer to the fetus.
Collapse
Affiliation(s)
- I Korchinski
- Women's Health Research Institute, Vancouver, Canada
| | - C Marquez
- British Columbia Center for Disease Control Public Health Laboratory, Vancouver, Canada
| | - E McClymont
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, Canada; Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - G Av-Gay
- Women's Health Research Institute, Vancouver, Canada; Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, Canada
| | - J Andrade
- Women's Health Research Institute, Vancouver, Canada
| | - C Elwood
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, Canada
| | - A Jassem
- British Columbia Center for Disease Control Public Health Laboratory, Vancouver, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - M Krajden
- British Columbia Center for Disease Control Public Health Laboratory, Vancouver, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - M Morshed
- British Columbia Center for Disease Control Public Health Laboratory, Vancouver, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - M Sadarangani
- Department of Pediatrics, University of British Columbia, Vancouver, Canada; Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, Canada
| | - G Tanunliong
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - I Sekirov
- British Columbia Center for Disease Control Public Health Laboratory, Vancouver, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - D Money
- Women's Health Research Institute, Vancouver, Canada; Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
3
|
Dombrowska-Pali A, Chrustek A, Olszewska-Słonina D, Socha MW. Composition and Antioxidant Status of Human Milk of Women Living in Bydgoszcz (Poland). Nutrients 2024; 16:3396. [PMID: 39408363 PMCID: PMC11479156 DOI: 10.3390/nu16193396] [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: 08/29/2024] [Revised: 09/27/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
OBJECTIVES The aim of this study was to compare cortisol concentrations, nutritional composition, and the antioxidant status of human milk of women living in Bydgoszcz (Poland), taking into account maternal factors (fertility, area of residence, economic activity, and breastfeeding period). METHODS The basic composition of human milk was evaluated using the MIRIS HMATM analyzer. The level of cortisol was determined by the enzyme-linked immunosorbent method. In order to determine the antioxidant activity, the DPPH radical method was used. RESULTS It was observed that the concentration of cortisol in human milk in the group of women living in the city center was higher compared to the milk of women living on the outskirts of the city. In the group of women breastfeeding from 3 to 5 weeks after childbirth, the concentration of cortisol in milk was higher compared to the group of women breastfeeding less than 12 months of age and compared to the group of women lactating over 12 months of age. The antioxidant status of human milk was highest in the group of professionally active women and in the group of breastfeeding women from 3 to 5 weeks after childbirth. The basic composition and the caloric value of human milk differed statistically significantly in the study groups. CONCLUSIONS Based on this study, it can be concluded that the composition and antioxidant status of human milk depends on maternal factors (fertility, professional activity, area of residence, and breastfeeding period). Higher cortisol concentrations in breast milk are probably determined by the area of residence (city center and associated higher noise/sound and stress levels) and lactation period (hormonal imbalance, fatigue, and postpartum period). Milk from economically active women shows greater protection against reactive oxygen species compared to milk from inactive women, protecting against the occurrence of diseases of civilization. Milk from breastfeeding women over 12 months of age also shows protection against reactive oxygen species, despite the fact that the highest level of antioxidant status of human milk occurs in the initial period of lactation.
Collapse
Affiliation(s)
- Agnieszka Dombrowska-Pali
- Department of Perinatology, Gynecology and Gynecologic Oncology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland
| | - Agnieszka Chrustek
- Department of Pathobiochemistry and Clinical Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie-Skłodowskiej 9, 85-094 Bydgoszcz, Poland
| | - Dorota Olszewska-Słonina
- Department of Pathobiochemistry and Clinical Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie-Skłodowskiej 9, 85-094 Bydgoszcz, Poland
| | - Maciej W. Socha
- Department of Perinatology, Gynecology and Gynecologic Oncology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland
- Department of Obstetrics and Gynecology, St. Adalberts’s Hospital in Gdańsk, Copernicus Healthcare Entity LLC, Jana Pawła II 50, 80-462 Gdańsk, Poland
| |
Collapse
|
4
|
Butler EA, Ray JG, Cohen E. Maternal-Newborn ABO Blood Groups and Risk of Bacterial Infection in Newborns. JAMA Netw Open 2024; 7:e2442227. [PMID: 39476233 PMCID: PMC11525604 DOI: 10.1001/jamanetworkopen.2024.42227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/08/2024] [Indexed: 11/02/2024] Open
Abstract
Importance Newborn immunity largely relies on maternal-fetal transfer of antibodies in utero. Incongruency in ABO blood groups between a mother and newborn may be associated with protection against serious infections, but data specific to newborn bacterial infections are lacking. Objective To ascertain the association between maternal-newborn ABO blood group incongruence and lower risk of bacterial infection in newborns. Design, Setting, and Participants This cohort study used linked patient-level datasets for all singleton live births between January 1, 2014, and December 31, 2020, in hospitals and health centers in Ontario, Canada. The cohort comprised maternal-newborn pairs with known ABO blood groups. Data analysis was conducted between February and May 2024. Exposure Maternal-newborn ABO blood group incongruence vs congruence. Main Outcomes and Measures The primary outcome was a bacterial infection arising in newborns within 30 days of birth. Bacterial infection was cultured from either blood, cerebrospinal fluid, urine, or lung specimen. Secondary outcomes were a bacterial infection with 7 days and 90 days of birth. Modified Poisson regression generated adjusted relative risks (ARRs) and 95% CIs, adjusted for neonatal sex and preterm birth. Results A total of 138 207 maternal-newborn pairs (maternal mean [SD] age, 31.8 [5.1] years among those with ABO blood group incongruency and 31.5 [5.1] years among those with ABO blood group congruency; newborn mean [SD] gestational age, 38.5 [2.3] weeks among those with incongruency and 38.4 [2.5] weeks among those with congruency; 19 475 males [51.3%] with incongruency and 52 041 males [51.9%] with congruency) were analyzed. Of these pairs, 37 953 (27.5%) had ABO blood group incongruency and 100 254 (72.5%) had ABO blood group congruency. Within 30 days of birth, 328 (8.6 per 1000) newborns in the incongruent group and 1029 (10.3 per 1000) newborns in the congruent group experienced a bacterial infection, corresponding to an ARR of 0.91 (95% CI, 0.81-1.03). The ARRs for bacterial infection within 7 days and 90 days of birth were 0.89 (95% CI, 0.73-1.09) and 0.86 (95% CI, 0.78-0.94), respectively. Conclusions and Relevance This cohort study found no association between maternal-newborn ABO blood group incongruence and risk of bacterial infection in newborns within 30 and 7 days of birth. However, incongruence was associated with a decreased risk of bacterial infection within 90 days of birth.
Collapse
Affiliation(s)
- Emily Ana Butler
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, Ontario, Canada
- ICES, Toronto, Ontario, Canada
| | - Joel G Ray
- ICES, Toronto, Ontario, Canada
- Department of Medicine, St Michael's Hospital, Toronto, Ontario, Canada
| | - Eyal Cohen
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, Ontario, Canada
- ICES, Toronto, Ontario, Canada
- Edwin S.H. Leong Centre for Healthy Children, Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Choi YJ, Lee DH, Song J, Kim KU, Min H, Chung SH, Kim TH, Kim CY, Kang I, Lee NM, Yi DY. Relationship of MicroRNA according to Immune Components of Breast Milk in Korean Lactating Mothers. Pediatr Gastroenterol Hepatol Nutr 2024; 27:322-331. [PMID: 39319280 PMCID: PMC11419785 DOI: 10.5223/pghn.2024.27.5.322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/05/2024] [Indexed: 09/26/2024] Open
Abstract
Purpose Human breast milk (HBM) contains immune components that produced and delivered from the mother along with nutrients necessary for the baby. MicroRNA (miRNA) is a small noncoding RNA molecule, that is used as an ideal biomarker for diagnosis and prognosis of various diseases and are more abundant in HBM. We analyzed and compared the immune components and miRNAs of HBM. Methods HBM were collected from 20 healthy breastfeeding mothers. We measured the amount of lactoferrin, lysozyme, and immunoglobulin A (IgA) and extracted the miRNAs from each breast milk samples. Next, the top 5 and bottom 5 expressed miRNAs were compared and analyzed based on the amounts of the 3 immune components. Results The mean levels and ranges of lactoferrin, lysozyme, and IgA were 6.33 (2.24-14.77)×106 ng/mL, 9.90 (1.42-17.59)×107 pg/mL, and 6.64 (0.48-20.01)×105 ng/mL, respectively. The miRNAs concentration per 1 mL of skim milk was 40.54 (14.95-110.01) ng/μL. Comparing the bottom 5 and top 5 groups of each immune component, 19 miRNAs were significantly upregulated (6, 9, and 4 targeting lactoferrin, lysozyme, and IgA, respectively) and 21 were significantly downregulated (4, 9, and 8 targeting lactoferrin, lysozyme, and IgA, respectively). There were no miRNAs that were expressed significantly higher or lower in common to all 3 components. However, 2 and 3 miRNAs were commonly overexpressed and underexpressed, in the top 5 groups of lysozyme and IgA concentrations. Conclusion We identified the immune components and miRNAs in breast milk and found that each individual has different ingredients.
Collapse
Affiliation(s)
- You Jin Choi
- Department of Pediatrics Inje University Ilsanpaik Hospital, Goyang, Korea
- College of Medicine, Chung-Ang University, Seoul, Korea
| | - Da Hye Lee
- Department of Pediatrics, Chung-Ang University Hospital, Seoul, Korea
| | - Jeonglyn Song
- Chung-Ang University Industry Academic Cooperation Foundation, Seoul, Korea
| | - Ki-Uk Kim
- College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Hyeyoung Min
- College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Sung-Hoon Chung
- Department of Pediatrics, Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Tae Hyeong Kim
- Department of Pediatrics, Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Chae-Young Kim
- Department of Pediatrics, Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Insoo Kang
- Section of Rheumatology, Allergy & Immunology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Na Mi Lee
- College of Medicine, Chung-Ang University, Seoul, Korea
- Department of Pediatrics, Chung-Ang University, Gwangmyeong Hospital, Gwangmyeong, Korea
| | - Dae Yong Yi
- College of Medicine, Chung-Ang University, Seoul, Korea
- Department of Pediatrics, Chung-Ang University Hospital, Seoul, Korea
| |
Collapse
|
6
|
Butler EA, Grandi SM, Matai L, Wang X, Cohen E, Ray JG. Differences in maternal-newborn ABO blood groups and risk of serious infant infection. QJM 2024; 117:512-519. [PMID: 38402542 PMCID: PMC11290255 DOI: 10.1093/qjmed/hcae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/11/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND During pregnancy, various maternal IgG antibodies are transferred to the developing fetus, some of which may protect the newborn against infection. If a mother and her fetus have different A, B or O (ABO) blood groups, then transferred maternal antibodies may plausibly protect the infant against infection. AIM To determine if maternal-newborn ABO blood group incongruence vs. congruence is associated with a lower risk of serious infection in the infant. DESIGN Retrospective population-based cohort. METHODS We used linked patient-level datasets for all singleton hospital livebirths from 2008 to 2022 in Ontario, Canada, with known maternal and newborn ABO blood groups. We used a dichotomous exposure state, either ABO blood group congruent (N = 114 507) or incongruent (N = 43 074). The main outcome of interest was the risk of serious infant infection within 27 days, and from 28 to 365 days, after birth. Cox proportional hazard models generated hazard ratios and 95% confidence intervals, and were adjusted for maternal age, world region of origin, residential income quintile and gestational age at birth. RESULTS Relative to maternal-newborn congruency, incongruent ABO blood group was associated with an adjusted hazard ratio of 0.88 (95% CI: 0.80-0.97) for serious neonatal infection within 27 days of birth, and 0.93 (95% CI: 0.90-0.96) for serious infection between 28 and 365 days after birth. CONCLUSIONS Maternal-newborn ABO incongruence may be associated with a lower relative risk of a serious infant infection within 27 days, and from 28 to 365 days, after birth.
Collapse
Affiliation(s)
- E A Butler
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, ON, Canada
- ICES, Toronto, ON, Canada
| | - S M Grandi
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, ON, Canada
- ICES, Toronto, ON, Canada
- Division of Epidemiology, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | | | - X Wang
- ICES, Toronto, ON, Canada
| | - E Cohen
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, ON, Canada
- ICES, Toronto, ON, Canada
| | - J G Ray
- ICES, Toronto, ON, Canada
- Department of Medicine, St Michael’s Hospital, Toronto, ON, Canada
| |
Collapse
|
7
|
Fukuda R, Pak K, Kiuchi M, Hirata N, Mochimaru N, Tanaka R, Mitsui M, Ohya Y, Yoshida K. Longitudinal Correlations between Molecular Compositions of Stratum Corneum and Breast Milk Factors during Infancy: A Prospective Birth Cohort Study. Nutrients 2024; 16:1897. [PMID: 38931252 PMCID: PMC11206726 DOI: 10.3390/nu16121897] [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: 05/01/2024] [Revised: 05/31/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Breast milk contains numerous factors that are involved in the maturation of the immune system and development of the gut microbiota in infants. These factors include transforming growth factor-β1 and 2, immunoglobin A, and lactoferrin. Breast milk factors may also affect epidermal differentiation and the stratum corneum (SC) barrier in infants, but no studies examining these associations over time during infancy have been reported. In this single-center exploratory study, we measured the molecular components of the SC using confocal Raman spectroscopy at 0, 1, 2, 6, and 12 months of age in 39 infants born at our hospital. Breast milk factor concentrations from their mothers' breast milk were determined. Correlation coefficients for the two datasets were estimated for each molecular component of the SC and breast milk factor at each age and SC depth. The results showed that breast milk factors and molecular components of the SC during infancy were partly correlated with infant age in months and SC depth, suggesting that breast milk factors influence the maturation of the SC components. These findings may improve understanding of the pathogenesis of skin diseases associated with skin barrier abnormalities.
Collapse
Affiliation(s)
- Risa Fukuda
- Division of Dermatology, National Center for Child Health and Development, Tokyo 157-8535, Japan; (R.F.); (R.T.)
| | - Kyongsun Pak
- Division of Biostatistics, Department of Data Management, Center of Clinical Research and Development, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Megumi Kiuchi
- Division of Research and Development, Pigeon Corporation, Ibaraki 300-2495, Japan
| | - Naoko Hirata
- Division of Research and Development, Pigeon Corporation, Ibaraki 300-2495, Japan
| | - Naoko Mochimaru
- Division of Dermatology, National Center for Child Health and Development, Tokyo 157-8535, Japan; (R.F.); (R.T.)
| | - Ryo Tanaka
- Division of Dermatology, National Center for Child Health and Development, Tokyo 157-8535, Japan; (R.F.); (R.T.)
| | - Mari Mitsui
- Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Yukihiro Ohya
- Allergy Center, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Kazue Yoshida
- Division of Dermatology, National Center for Child Health and Development, Tokyo 157-8535, Japan; (R.F.); (R.T.)
- Allergy Center, National Center for Child Health and Development, Tokyo 157-8535, Japan
| |
Collapse
|
8
|
Eker F, Akdaşçi E, Duman H, Yalçıntaş YM, Canbolat AA, Kalkan AE, Karav S, Šamec D. Antimicrobial Properties of Colostrum and Milk. Antibiotics (Basel) 2024; 13:251. [PMID: 38534686 DOI: 10.3390/antibiotics13030251] [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: 01/31/2024] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024] Open
Abstract
The growing number of antibiotic resistance genes is putting a strain on the ecosystem and harming human health. In addition, consumers have developed a cautious attitude towards chemical preservatives. Colostrum and milk are excellent sources of antibacterial components that help to strengthen the immunity of the offspring and accelerate the maturation of the immune system. It is possible to study these important defenses of milk and colostrum, such as lactoferrin, lysozyme, immunoglobulins, oligosaccharides, etc., as biotherapeutic agents for the prevention and treatment of numerous infections caused by microbes. Each of these components has different mechanisms and interactions in various places. The compound's mechanisms of action determine where the antibacterial activity appears. The activation of the antibacterial activity of milk and colostrum compounds can start in the infant's mouth during lactation and continue in the gastrointestinal regions. These antibacterial properties possess potential for therapeutic uses. In order to discover new perspectives and methods for the treatment of bacterial infections, additional investigations of the mechanisms of action and potential complexes are required.
Collapse
Affiliation(s)
- Furkan Eker
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Emir Akdaşçi
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Hatice Duman
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Yalçın Mert Yalçıntaş
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Ahmet Alperen Canbolat
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Arda Erkan Kalkan
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Dunja Šamec
- Department of Food Technology, University North, Trg Dr. Žarka Dolinara 1, 48000 Koprivnica, Croatia
| |
Collapse
|
9
|
Parker LA, Koernere R, Fordham K, Bubshait H, Eugene A, Gefre A, Bendixen M. Mother's Own Milk Versus Donor Human Milk: What's the Difference? Crit Care Nurs Clin North Am 2024; 36:119-133. [PMID: 38296370 DOI: 10.1016/j.cnc.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Mother's own milk (MOM) is known to decrease complications in preterm infants and when unavailable, it is recommended that preterm very low-birth weight infants be fed donor human milk (DHM). Due to the pasteurization, processing, and lactation stage of donors, DHM does not contain the same nutritional, immunologic, and microbial components as MOM. This review summarizes the differences between MOM and DHM, the potential effects on health outcomes, and the clinical implications of these differences. Finally, implications for research and clinical practice are discussed.
Collapse
Affiliation(s)
- Leslie A Parker
- College of Nursing, University of Florida, Box 100187 College of Nursing, Gainesville, FL, USA.
| | - Rebecca Koernere
- College of Nursing, University of Florida, Box 100187 College of Nursing, Gainesville, FL, USA
| | - Keliy Fordham
- College of Nursing, University of Florida, Box 100187 College of Nursing, Gainesville, FL, USA
| | - Hussah Bubshait
- College of Nursing, University of Florida, Box 100187 College of Nursing, Gainesville, FL, USA
| | - Alissandre Eugene
- College of Nursing, University of Florida, Box 100187 College of Nursing, Gainesville, FL, USA
| | - Adrienne Gefre
- College of Nursing, University of Florida, Box 100187 College of Nursing, Gainesville, FL, USA
| | - Marion Bendixen
- College of Nursing, University of Florida, Box 100187 College of Nursing, Gainesville, FL, USA
| |
Collapse
|
10
|
Valcarce V, Stafford LS, Neu J, Parker L, Vicuna V, Cross T, D'Agati O, Diakite S, Haley A, Feigenbaum J, Al Mahmoud MY, Visvalingam A, Cacho N, Kosik I, Yewdell JW, Larkin J. COVID-19 booster enhances IgG mediated viral neutralization by human milk in vitro. Front Nutr 2024; 11:1289413. [PMID: 38406184 PMCID: PMC10884187 DOI: 10.3389/fnut.2024.1289413] [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: 09/06/2023] [Accepted: 01/25/2024] [Indexed: 02/27/2024] Open
Abstract
Background Facilitated by the inability to vaccinate, and an immature immune system, COVID-19 remains a leading cause of death among children. Vaccinated lactating mothers produce specific SARS-CoV-2 antibodies in their milk, capable of neutralizing the virus in vitro. Our objective for this study is to assess the effect of COVID-19 booster dose on SARS-CoV-2 antibody concentration and viral neutralization in milk, plasma, and infant stool. Methods Thirty-nine mothers and 25 infants were enrolled from December 2020 to May 2022. Milk, maternal plasma, and infants' stool were collected at various time-points up to 12 months following mRNA COVID-19 vaccination. A subgroup of 14 mothers received a booster dose. SARS-CoV-2 antibody levels and their neutralization capacities were assessed. Results Booster vaccination led to significantly higher IgG levels within human milk and breastfed infants' stool. In vitro neutralization of VSV-gfp-SARS-CoV-2-S-gp, a laboratory safe SARS-CoV-2 like pseudovirus, improved following the booster, with a 90% increase in plasma neutralization and a 60% increase in milk neutralization. We found that post-booster neutralization by human milk was highly correlated to SARS-CoV-2 IgG level. In support of our correlation result, Protein G column depletion of IgG in milk yielded a significant reduction in viral neutralization (p = 0.04). Discussion The substantial increase in neutralizing IgG levels in milk and breastfed infants' stool post-booster, coupled with the decrease in milk neutralization capabilities upon IgG depletion, underscores the efficacy of booster doses in augmenting the immune response against SARS-CoV-2 in human milk.
Collapse
Affiliation(s)
- Vivian Valcarce
- Department of Pediatrics, University of Florida, Gainesville, FL, United States
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Lauren Stewart Stafford
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, United States
| | - Josef Neu
- Department of Pediatrics, University of Florida, Gainesville, FL, United States
| | - Leslie Parker
- College of Nursing, University of Florida, Gainesville, FL, United States
| | - Valeria Vicuna
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, United States
| | - Tyler Cross
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, United States
| | - Olivia D'Agati
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, United States
| | - Sisse Diakite
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, United States
| | - Addison Haley
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, United States
| | - Jake Feigenbaum
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, United States
| | - Mahmoud Y. Al Mahmoud
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, United States
| | - Anjali Visvalingam
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, United States
| | - Nicole Cacho
- Department of Pediatrics, University of Florida, Gainesville, FL, United States
- Department of Pediatrics, University of California Davis, Sacramento, CA, United States
| | - Ivan Kosik
- Laboratory of Viral Diseases, National Institutes of Health, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Jonathan W. Yewdell
- Laboratory of Viral Diseases, National Institutes of Health, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Joseph Larkin
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, United States
| |
Collapse
|
11
|
de Graaf SC, Bondt A, van Rijswijck DMH, Juncker HG, Mulleners SJ, Damen MJA, Hoek M, van Keulen BJ, van Goudoever JB, Heck AJR, Dingess KA. A case series exploring the human milk polyclonal IgA1 response to repeated SARS-CoV-2 vaccinations by LC-MS based fab profiling. Front Nutr 2024; 10:1305086. [PMID: 38288064 PMCID: PMC10822949 DOI: 10.3389/fnut.2023.1305086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/20/2023] [Indexed: 01/31/2024] Open
Abstract
Introduction Upon vaccination against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) humans will start to produce antibodies targeting virus specific antigens that will end up in circulation. In lactating women such antibodies will also end up in breastmilk, primarily in the form of secretory immunoglobulin A1 (SIgA1), the most abundant immunoglobulin (Ig) in human milk. Here we set out to investigate the SIgA1 clonal repertoire response to repeated SARS-CoV-2 vaccination, using a LC-MS fragment antigen-binding (Fab) clonal profiling approach. Methods We analyzed the breastmilk of six donors from a larger cohort of 109 lactating mothers who received one of three commonly used SARS-CoV-2 vaccines. We quantitatively monitored the SIgA1 Fab clonal profile over 16 timepoints, from just prior to the first vaccination until 15 days after the second vaccination. Results In all donors, we detected a population of 89-191 vaccine induced clones. These populations were unique to each donor and heterogeneous with respect to individual clonal concentrations, total clonal titer, and population size. The vaccine induced clones were dominated by persistent clones (68%) which came up after the first vaccination and were retained or reoccurred after the second vaccination. However, we also observe transient SIgA1 clones (16%) which dissipated before the second vaccination, and vaccine induced clones which uniquely emerged only after the second vaccination (16%). These distinct populations were observed in all analyzed donors, regardless of the administered vaccine. Discussion Our findings suggest that while individual donors have highly unique human milk SIgA1 clonal profiles and a highly personalized SIgA1 response to SARS-CoV-2 vaccination, there are also commonalities in vaccine induced responses.
Collapse
Affiliation(s)
- Sebastiaan C. de Graaf
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Albert Bondt
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Danique M. H. van Rijswijck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Hannah G. Juncker
- Department of Pediatrics, Amsterdam Reproduction and Development Research Institute, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, Netherlands
| | - Sien J. Mulleners
- Department of Pediatrics, Amsterdam Reproduction and Development Research Institute, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Mirjam J. A. Damen
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Max Hoek
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Britt J. van Keulen
- Department of Pediatrics, Amsterdam Reproduction and Development Research Institute, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Johannes B. van Goudoever
- Department of Pediatrics, Amsterdam Reproduction and Development Research Institute, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Albert J. R. Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Kelly A. Dingess
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
- Department of Pediatrics, Amsterdam Reproduction and Development Research Institute, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
12
|
Hochmayr C, Toferer M, Hammerl M, Winkler I, Huber E, Urbanek M, Kiechl-Kohlendorfer U, Griesmaier E, Posod A. Breast milk anti-S1RBD immunoglobulin concentrations throughout lactation: An observational report. Acta Paediatr 2024; 113:81-83. [PMID: 37814794 DOI: 10.1111/apa.16997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 08/30/2023] [Accepted: 10/03/2023] [Indexed: 10/11/2023]
Affiliation(s)
- Christoph Hochmayr
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Innsbruck, Austria
| | - Martha Toferer
- Department of Obstetrics and Gynecology, Kardinal Schwarzenberg Hospital, Schwarzach im Pongau, Austria
| | - Marlene Hammerl
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Innsbruck, Austria
| | - Ira Winkler
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Innsbruck, Austria
| | - Eva Huber
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Innsbruck, Austria
| | - Martina Urbanek
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Innsbruck, Austria
| | | | - Elke Griesmaier
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Innsbruck, Austria
| | - Anna Posod
- Department of Pediatrics II (Neonatology), Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
13
|
Sereme Y, Toumi E, Saifi E, Faury H, Skurnik D. Maternal immune factors involved in the prevention or facilitation of neonatal bacterial infections. Cell Immunol 2024; 395-396:104796. [PMID: 38104514 DOI: 10.1016/j.cellimm.2023.104796] [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: 10/22/2023] [Revised: 11/30/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
Newborns, whether born prematurely or at term, have a fully formed but naive immune system that must adapt to the extra-uterine environment to prevent infections. Maternal immunity, transmitted through the placenta and breast milk, protects newborns against infections, primarily via immunoglobulins (IgG and IgA) and certain maternal immune cells also known as microchimeric cells. Recently, it also appeared that the maternal gut microbiota played a vital role in neonatal immune maturation via microbial compounds impacting immune development and the establishment of immune tolerance. In this context, maternal vaccination is a powerful tool to enhance even more maternal and neonatal health. It involves the transfer of vaccine-induced antibodies to protect both mother and child from infectious diseases. In this work we review the state of the art on maternal immune factors involved in the prevention of neonatal bacterial infections, with particular emphasis on the role of maternal vaccination in protecting neonates against bacterial disease.
Collapse
Affiliation(s)
- Youssouf Sereme
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Eya Toumi
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Estelle Saifi
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Helène Faury
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades, F-75015 Paris, France; Department of Microbiology, Necker Hospital, University de Paris, Paris, France
| | - David Skurnik
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades, F-75015 Paris, France; Department of Microbiology, Necker Hospital, University de Paris, Paris, France; FHU PREMA, Paris, France.
| |
Collapse
|
14
|
Oskam N, den Boer MA, Lukassen MV, Ooijevaar-de Heer P, Veth TS, van Mierlo G, Lai SH, Derksen NIL, Yin V, Streutker M, Franc V, Šiborová M, Damen MJA, Kos D, Barendregt A, Bondt A, van Goudoever JB, de Haas CJC, Aerts PC, Muts RM, Rooijakkers SHM, Vidarsson G, Rispens T, Heck AJR. CD5L is a canonical component of circulatory IgM. Proc Natl Acad Sci U S A 2023; 120:e2311265120. [PMID: 38055740 PMCID: PMC10723121 DOI: 10.1073/pnas.2311265120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 11/07/2023] [Indexed: 12/08/2023] Open
Abstract
Immunoglobulin M (IgM) is an evolutionary conserved key component of humoral immunity, and the first antibody isotype to emerge during an immune response. IgM is a large (1 MDa), multimeric protein, for which both hexameric and pentameric structures have been described, the latter additionally containing a joining (J) chain. Using a combination of single-particle mass spectrometry and mass photometry, proteomics, and immunochemical assays, we here demonstrate that circulatory (serum) IgM exclusively exists as a complex of J-chain-containing pentamers covalently bound to the small (36 kDa) protein CD5 antigen-like (CD5L, also called apoptosis inhibitor of macrophage). In sharp contrast, secretory IgM in saliva and milk is principally devoid of CD5L. Unlike IgM itself, CD5L is not produced by B cells, implying that it associates with IgM in the extracellular space. We demonstrate that CD5L integration has functional implications, i.e., it diminishes IgM binding to two of its receptors, the FcαµR and the polymeric Immunoglobulin receptor. On the other hand, binding to FcµR as well as complement activation via C1q seem unaffected by CD5L integration. Taken together, we redefine the composition of circulatory IgM as a J-chain containing pentamer, always in complex with CD5L.
Collapse
Affiliation(s)
- Nienke Oskam
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Amsterdam University Medical Center, Amsterdam1066 CX, the Netherlands
| | - Maurits A. den Boer
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht3584 CH, the Netherlands
- Netherlands Proteomics Center, Utrecht3584 CH, the Netherlands
| | - Marie V. Lukassen
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht3584 CH, the Netherlands
- Netherlands Proteomics Center, Utrecht3584 CH, the Netherlands
| | - Pleuni Ooijevaar-de Heer
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Amsterdam University Medical Center, Amsterdam1066 CX, the Netherlands
| | - Tim S. Veth
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht3584 CH, the Netherlands
- Netherlands Proteomics Center, Utrecht3584 CH, the Netherlands
| | - Gerard van Mierlo
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Amsterdam University Medical Center, Amsterdam1066 CX, the Netherlands
| | - Szu-Hsueh Lai
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht3584 CH, the Netherlands
- Netherlands Proteomics Center, Utrecht3584 CH, the Netherlands
| | - Ninotska I. L. Derksen
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Amsterdam University Medical Center, Amsterdam1066 CX, the Netherlands
| | - Victor Yin
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht3584 CH, the Netherlands
- Netherlands Proteomics Center, Utrecht3584 CH, the Netherlands
| | - Marij Streutker
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Amsterdam University Medical Center, Amsterdam1066 CX, the Netherlands
| | - Vojtech Franc
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht3584 CH, the Netherlands
- Netherlands Proteomics Center, Utrecht3584 CH, the Netherlands
| | - Marta Šiborová
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht3584 CH, the Netherlands
- Netherlands Proteomics Center, Utrecht3584 CH, the Netherlands
| | - Mirjam J. A. Damen
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht3584 CH, the Netherlands
- Netherlands Proteomics Center, Utrecht3584 CH, the Netherlands
| | - Dorien Kos
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Amsterdam University Medical Center, Amsterdam1066 CX, the Netherlands
| | - Arjan Barendregt
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht3584 CH, the Netherlands
- Netherlands Proteomics Center, Utrecht3584 CH, the Netherlands
| | - Albert Bondt
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht3584 CH, the Netherlands
- Netherlands Proteomics Center, Utrecht3584 CH, the Netherlands
| | - Johannes B. van Goudoever
- Amsterdam University Medical Center, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam1105 AZ, the Netherlands
| | - Carla J. C. de Haas
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht3584 CX, the Netherlands
| | - Piet C. Aerts
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht3584 CX, the Netherlands
| | - Remy M. Muts
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht3584 CX, the Netherlands
| | - Suzan H. M. Rooijakkers
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht3584 CX, the Netherlands
| | - Gestur Vidarsson
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht3584 CH, the Netherlands
- Netherlands Proteomics Center, Utrecht3584 CH, the Netherlands
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunohematology, Amsterdam University Medical Center, Amsterdam1066 CX, the Netherlands
| | - Theo Rispens
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Amsterdam University Medical Center, Amsterdam1066 CX, the Netherlands
| | - Albert J. R. Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht3584 CH, the Netherlands
- Netherlands Proteomics Center, Utrecht3584 CH, the Netherlands
| |
Collapse
|
15
|
Abels E, Jacobs JW, Prior D, Willets LC, Sostin N, Tormey CA, Binns TC. Passive transfer of alloantibodies through breast milk as a mediator of hemolytic anemia. Transfusion 2023; 63:2188-2196. [PMID: 37706556 DOI: 10.1111/trf.17548] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/21/2023] [Accepted: 08/01/2023] [Indexed: 09/15/2023]
Abstract
BACKGROUND Hemolytic disease of the fetus and newborn (HDFN) is characterized by destruction of fetal/neonatal red blood cells (RBCs) secondary to maternally derived antibodies, which are typically thought to be passively acquired via placental transfer. Few cases have examined the possibility of HDFN mediated by maternal antibodies passively transferred via breast milk. METHODS We describe two cases of persistent HDFN in infants potentially mediated by passively acquired antibodies via maternal breast milk. We discuss supporting and refuting evidence that may account for this possibility and describe testing methodology illustrating how maternal alloantibodies can be detected in breast milk. RESULTS In both cases, anti-D antibodies were detected in maternal breast milk. One patient experienced a significant decrease in anti-D plasma titer from 64 to 4 dilutions following 2 weeks of breastfeeding cessation. The other patient experienced a resolution of anemia without breastfeeding cessation. CONCLUSION There is a paucity of data regarding the lifespan of passively acquired RBC antibodies in neonatal circulation, with significant variation noted between passively acquired IgG based on studies utilizing intravenous immunoglobulin compared to studies of maternally-acquired antiviral IgG antibodies. While our data do not definitively implicate passive transfer of alloantibodies in breast milk as a mediator of HDFN, they do illustrate the need for further investigation into the mechanisms and kinetics of passively acquired antibodies in neonatal circulation.
Collapse
Affiliation(s)
- Elizabeth Abels
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jeremy W Jacobs
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Daniel Prior
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Laura C Willets
- Pediatric Clinical Nutrition, Yale New Haven Hospital, New Haven, Connecticut, USA
| | - Nataliya Sostin
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Christopher A Tormey
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Thomas C Binns
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
16
|
Caffé B, Blackwell A, Fehrenkamp BD, Williams JE, Pace RM, Lackey KA, Ruiz L, Rodríguez JM, McGuire MA, Foster JA, Sellen DW, Kamau-Mbuthia EW, Kamundia EW, Mbugua S, Moore SE, Prentice AM, Kvist LJ, Otoo GE, Pareja RG, Bode L, Gebeyehu D, Gindola DK, Boothman S, Flores K, McGuire MK, Meehan CL. Human milk immune factors, maternal nutritional status, and infant sex: The INSPIRE study. Am J Hum Biol 2023; 35:e23943. [PMID: 37358306 PMCID: PMC10749986 DOI: 10.1002/ajhb.23943] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 05/24/2023] [Accepted: 06/08/2023] [Indexed: 06/27/2023] Open
Abstract
OBJECTIVES Breastfeeding is an energetically costly and intense form of human parental investment, providing sole-source nutrition in early infancy and bioactive components, including immune factors. Given the energetic cost of lactation, milk factors may be subject to tradeoffs, and variation in concentrations have been explored utilizing the Trivers-Willard hypothesis. As human milk immune factors are critical to developing immune system and protect infants against pathogens, we tested whether concentrations of milk immune factors (IgA, IgM, IgG, EGF, TGFβ2, and IL-10) vary in response to infant sex and maternal condition (proxied by maternal diet diversity [DD] and body mass index [BMI]) as posited in the Trivers-Willard hypothesis and consider the application of the hypothesis to milk composition. METHODS We analyzed concentrations of immune factors in 358 milk samples collected from women residing in 10 international sites using linear mixed-effects models to test for an interaction between maternal condition, including population as a random effect and infant age and maternal age as fixed effects. RESULTS IgG concentrations were significantly lower in milk produced by women consuming diets with low diversity with male infants than those with female infants. No other significant associations were identified. CONCLUSIONS IgG concentrations were related to infant sex and maternal diet diversity, providing minimal support for the hypothesis. Given the lack of associations across other select immune factors, results suggest that the Trivers-Willard hypothesis may not be broadly applied to human milk immune factors as a measure of maternal investment, which are likely buffered against perturbations in maternal condition.
Collapse
Affiliation(s)
- Beatrice Caffé
- Department of Anthropology, Washington State University, Pullman, Washington, USA
| | - Aaron Blackwell
- Department of Anthropology, Washington State University, Pullman, Washington, USA
| | - Bethaney D Fehrenkamp
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, Idaho, USA
- Washington, Wyoming, Alaska, Montana, Idaho (WWAMI) Medical Education Program, University of Idaho, Moscow, Idaho, USA
| | - Janet E Williams
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, Idaho, USA
| | - Ryan M Pace
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, Idaho, USA
| | - Kimberly A Lackey
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, Idaho, USA
| | - Lorena Ruiz
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Microhealth Group, Oviedo, Spain
| | - Juan M Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Mark A McGuire
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, Idaho, USA
| | - James A Foster
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, USA
| | - Daniel W Sellen
- Department of Anthropology, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Samwel Mbugua
- Department of Human Nutrition, Egerton University, Nakuru, Kenya
| | - Sophie E Moore
- Department of Women and Children's Health, King's College London, London, UK
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Andrew M Prentice
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | | | - Gloria E Otoo
- Department of Nutrition and Food Science, University of Ghana, Accra, Ghana
| | | | - Lars Bode
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California, San Diego, La Jolla, California, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Dubale Gebeyehu
- Department of Anthropology, Hawassa University, Hawassa, Ethiopia
| | - Debela K Gindola
- Department of Anthropology, Hawassa University, Hawassa, Ethiopia
| | - Sarah Boothman
- School of Biological Sciences, Washington State University, Pullman, Washington, USA
| | - Katherine Flores
- Department of Anthropology, Washington State University, Pullman, Washington, USA
| | - Michelle K McGuire
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, Idaho, USA
| | - Courtney L Meehan
- Department of Anthropology, Washington State University, Pullman, Washington, USA
| |
Collapse
|
17
|
Enyoh CE, Ovuoraye PE, Qingyue W, Wang W. Examining the impact of nanoplastics and PFAS exposure on immune functions through inhibition of secretory immunoglobin A in human breast milk. JOURNAL OF HAZARDOUS MATERIALS 2023; 459:132103. [PMID: 37527590 DOI: 10.1016/j.jhazmat.2023.132103] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/18/2023] [Accepted: 07/18/2023] [Indexed: 08/03/2023]
Abstract
Emerging contaminants such as nanoplastics (NPs) and per- and polyfluoroalkyl substances (PFAS), have been detected in the environment and breast milk, thus exposing infants to potentially harmful chemicals during breastfeeding. Breast milk contains secretory immunoglobulin A (SIgA), an antibody that plays a vital role in disease protection and the development of the infant's immune system. This study employed molecular simulation and fractional factorial designs to assess the toxicity of NPs and PFAS on breast milk and their influence on infant immunity by inhibiting SIgA. The research found that NPs and PFAS have higher binding affinities to SIgA compared to the control compound. Polycarbonate (-10.7 kcal/mol) had the highest binding affinity among plastics, while Perfluorodecanoic acid (PFDA, - 8.0 kcal/mol) had the highest binding affinity among PFAS. The relative toxic index was higher for PFAS (2.4) than for plastics (1.9), suggesting that PFAS may pose a higher overall toxicity burden on the protein. The presence of specific combinations of NPs and PFAS in breast milk may potentially harm breastfeeding infants, although additional experimental studies are required to validate these findings. These results underscore the potential risks associated with these emerging contaminants in breast milk and their impact on infant immunity.
Collapse
Affiliation(s)
- Christian Ebere Enyoh
- Graduate School of Science and Engineering, Saitama University, 255 Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan.
| | - Prosper E Ovuoraye
- Department of Chemical Engineering, Federal University of Petroleum Resources, PMB 1221 Effurun, Nigeria
| | - Wang Qingyue
- Graduate School of Science and Engineering, Saitama University, 255 Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan.
| | - Weiqian Wang
- Graduate School of Science and Engineering, Saitama University, 255 Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan
| |
Collapse
|
18
|
Arishi RA, Lai CT, Geddes DT, Stinson LF. Impact of breastfeeding and other early-life factors on the development of the oral microbiome. Front Microbiol 2023; 14:1236601. [PMID: 37744908 PMCID: PMC10513450 DOI: 10.3389/fmicb.2023.1236601] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/25/2023] [Indexed: 09/26/2023] Open
Abstract
The oral cavity is home to the second most diverse microbiome in the human body. This community contributes to both oral and systemic health. Acquisition and development of the oral microbiome is a dynamic process that occurs over early life; however, data regarding longitudinal assembly of the infant oral microbiome is scarce. While numerous factors have been associated with the composition of the infant oral microbiome, early feeding practices (breastfeeding and the introduction of solids) appear to be the strongest determinants of the infant oral microbiome. In the present review, we draw together data on the maternal, infant, and environmental factors linked to the composition of the infant oral microbiome, with a focus on early nutrition. Given evidence that breastfeeding powerfully shapes the infant oral microbiome, the review explores potential mechanisms through which human milk components, including microbes, metabolites, oligosaccharides, and antimicrobial proteins, may interact with and shape the infant oral microbiome. Infancy is a unique period for the oral microbiome. By enhancing our understanding of oral microbiome assembly in early life, we may better support both oral and systemic health throughout the lifespan.
Collapse
Affiliation(s)
- Roaa A. Arishi
- School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia
- Ministry of Health, Riyadh, Saudi Arabia
| | - Ching T. Lai
- School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia
| | - Donna T. Geddes
- School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia
| | - Lisa F. Stinson
- School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
19
|
Petersson M, Thrane SW, Gram L, Muyldermans S, Laustsen AH. Orally delivered single-domain antibodies against gastrointestinal pathogens. Trends Biotechnol 2023; 41:875-886. [PMID: 36774206 DOI: 10.1016/j.tibtech.2023.01.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 01/13/2023] [Accepted: 01/20/2023] [Indexed: 02/11/2023]
Abstract
Single-domain antibodies (sdAbs) are exceptionally stable fragments derived from the antigen-binding domains of immunoglobulins. They can withstand extreme pH, high temperature, and proteolysis, making them suitable for controlling gastrointestinal (GI) infections in humans and animals. sdAbs may function in their native soluble form, although different derived protein formats and the use of delivery vehicles can be useful for improved oral delivery. We discuss selected examples of the use of orally delivered sdAbs for protecting humans and animals against GI infections caused by pathogenic bacteria, viruses, and parasites. We finally provide perspectives on how sdAbs may be applied industrially and what challenges should be overcome for orally delivered sdAbs to reach the market.
Collapse
Affiliation(s)
- Marcus Petersson
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark; Bactolife A/S, Copenhagen East, Denmark
| | | | - Lone Gram
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Serge Muyldermans
- Department of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Andreas H Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark; Bactolife A/S, Copenhagen East, Denmark.
| |
Collapse
|
20
|
Chanda BM, Chen XQ. Breastfeeding during the COVID-19 pandemic. Front Pediatr 2023; 11:1120763. [PMID: 37342530 PMCID: PMC10277472 DOI: 10.3389/fped.2023.1120763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 05/10/2023] [Indexed: 06/23/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused many significant changes to all aspects of day to day life. The disease has spread and reached pandemic proportions. The principle route of transmission is the respiratory route. Infants, pregnant women and breastfeeding mothers have all been affected. Many interventions and guidelines from important societies have been instituted in order to curb the transmission of the disease. These have involved both pharmacological and non-pharmacological methods. COVID-19 vaccines have also emerged as important methods of primary prevention of the disease. But several questions have been raised concerning the safety and efficacy of their use in pregnant and breastfeeding mothers. It has also not been clear if the vaccines are effective in generating a robust immune response in the pregnant women and breastfeeding mothers to confer passive immunity to the fetuses and infants, respectively. And they have not been tested in infants. The aspect of infant feeding has equally been affected. Although breast milk has not been known to serve as the vehicle of transmission of the virus, there is still some lack of uniformity of practice regarding breastfeeding when a mother has SARS-CoV-2 infection. This has led to infant feeding being done by the use of commercial formula feeds, pasteurized human donor breast milk, feeding on the mother's own expressed breast milk by a care giver and directly breastfeeding with skin to skin contact. This is despite breast milk being the most physiologically appropriate type of feed for infants. Therefore the pertinent question remains; should breastfeeding continue during the pandemic continue? This review also seeks to analyse the vast amount of scientific information regarding the subject and to synthesize science-based information.
Collapse
Affiliation(s)
- Bwalya Mpelwa Chanda
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao-Qing Chen
- Department of Pediatrics, First Affiliation Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
21
|
Elbeltagi R, Al-Beltagi M, Saeed NK, Bediwy AS. Cardiometabolic effects of breastfeeding on infants of diabetic mothers. World J Diabetes 2023; 14:617-631. [PMID: 37273257 PMCID: PMC10236993 DOI: 10.4239/wjd.v14.i5.617] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/01/2023] [Accepted: 04/07/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Breast milk is the best and principal nutritional source for neonates and infants. It may protect infants against many metabolic diseases, predominantly obesity and type 2 diabetes. Diabetes mellitus (DM) is a chronic metabolic and microvascular disease that affects all the body systems and all ages from intrauterine life to late adulthood. Breastfeeding protects against infant mortality and diseases, such as necrotizing enterocolitis, diarrhoea, respiratory infections, viral and bacterial infection, eczema, allergic rhinitis, asthma, food allergies, malocclusion, dental caries, Crohn's disease, and ulcerative colitis. It also protects against obesity and insulin resistance and increases intelligence and mental development. Gestational diabetes has short and long-term impacts on infants of diabetic mothers (IDM). Breast milk composition changes in mothers with gestational diabetes.
AIM To investigate the beneficial or detrimental effects of breastfeeding on the cardiometabolic health of IDM and their mothers.
METHODS We performed a database search on different engines and a thorough literature review and included 121 research published in English between January 2000 and December 15, 2022, in this review.
RESULTS Most of the literature agreed on the beneficial effects of breast milk for both the mother and the infant in the short and long terms. Breastfeeding protects mothers with gestational diabetes against obesity and type 2 DM. Despite some evidence of the protective effects of breastfeeding on IDM in the short and long term, the evidence is not strong enough due to the presence of many confounding factors and a lack of sufficient studies.
CONCLUSION We need more comprehensive research to prove these effects. Despite many obstacles that may enface mothers with gestational diabetes to start and maintain breastfeeding, every effort should be made to encourage them to breastfeed.
Collapse
Affiliation(s)
- Reem Elbeltagi
- Department of Medicine, Irish Royal College of Surgeon, Busaiteen 15503, Bahrain
| | - Mohammed Al-Beltagi
- Department of Pediatrics, Faculty of Medicine, Tanta University, Tanta 31511, Egypt
- Department of Pediatrics, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
| | - Nermin Kamal Saeed
- Medical Microbiology Section, Department of Pathology, Salmaniya Medical Complex, Ministry of Health, Kingdom of Bahrain, Manama 12, Bahrain
- Department of Microbiology, Irish Royal College of Surgeon, Bahrain, Busaiteen 15503, Bahrain
| | - Adel Salah Bediwy
- Department of Chest Diseases, Faculty of Medicine, Tanta University, Tanta 31527, Egypt
- Department of Pulmonology, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
| |
Collapse
|
22
|
Cortés-Sarabia K, Guzman-Silva V, Martinez-Pacheco KM, Meza-Hernández JA, Luna-Pineda VM, Leyva-Vázquez MA, Vences-Velázquez A, Beltrán-Anaya FO, Del Moral-Hernández O, Illades-Aguiar B. Detection of IgA and IgG Antibodies against the Structural Proteins of SARS-CoV-2 in Breast Milk and Serum Samples Derived from Breastfeeding Mothers. Viruses 2023; 15:966. [PMID: 37112946 PMCID: PMC10144911 DOI: 10.3390/v15040966] [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: 02/28/2023] [Revised: 04/03/2023] [Accepted: 04/09/2023] [Indexed: 04/29/2023] Open
Abstract
Background: COVID-19 vaccination or natural infection is associated with the development of immunity. The search of IgA and IgG antibodies against all the structural proteins (spike, nucleocapsid, membrane, and envelope) of SARS-CoV-2 in breastfeeding mothers is associated with immunity that can help the newborn avoid development of the infection. Methods: In this study, we analyzed 30 breastfeeding women that provided samples of breast milk and serum and evaluated the presence of IgA, total IgG, and subclasses against the structural proteins of SARS-CoV-2. Results: We reported a high seroprevalence to IgA (76.67-100%) and negativity to IgG against all analyzed proteins in breast milk. Seroprevalence in serum samples was around 10-36.67% to IgA and 23.3-60% to IgG. Finally, we detected the presence of the subclasses IgG1, IgG2, and IgG4 against all the structural proteins of SARS-CoV-2. Conclusions: This work provides evidence of the presence of IgA and IgG antibodies against the four structural proteins of SARS-CoV-2 in breast milk and serum samples derived from breastfeeding women, which can confer immunity to the newborn.
Collapse
Affiliation(s)
- Karen Cortés-Sarabia
- Laboratorio de Inmunobiología y Diagnóstico Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39086, Mexico
| | - Vianey Guzman-Silva
- Laboratorio de Inmunobiología y Diagnóstico Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39086, Mexico
| | - Karla Montserrat Martinez-Pacheco
- Laboratorio de Inmunobiología y Diagnóstico Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39086, Mexico
| | - Jesús Alberto Meza-Hernández
- Laboratorio de Inmunobiología y Diagnóstico Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39086, Mexico
| | - Víctor Manuel Luna-Pineda
- Unidad de Investigación en Inmunología y Proteómica, Laboratorio de Investigación en COVID-19, Hospital Infantil de México “Federico Gómez”, Mexico City 06720, Mexico
| | - Marco Antonio Leyva-Vázquez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39086, Mexico
| | - Amalia Vences-Velázquez
- Laboratorio de Inmunobiología y Diagnóstico Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39086, Mexico
| | - Fredy Omar Beltrán-Anaya
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39086, Mexico
| | - Oscar Del Moral-Hernández
- Laboratorio de Virología, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39086, Mexico
| | - Berenice Illades-Aguiar
- Unidad de Investigación en Inmunología y Proteómica, Laboratorio de Investigación en COVID-19, Hospital Infantil de México “Federico Gómez”, Mexico City 06720, Mexico
| |
Collapse
|
23
|
Schulz M, Levy DI, Petropoulos CJ, Bashirians G, Winburn I, Mahn M, Somanathan S, Cheng SH, Byrne BJ. Binding and neutralizing anti-AAV antibodies: Detection and implications for rAAV-mediated gene therapy. Mol Ther 2023; 31:616-630. [PMID: 36635967 PMCID: PMC10014285 DOI: 10.1016/j.ymthe.2023.01.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/21/2022] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
Assessment of anti-adeno-associated virus (AAV) antibodies in patients prior to systemic gene therapy administration is an important consideration regarding efficacy and safety of the therapy. Approximately 30%-60% of individuals have pre-existing anti-AAV antibodies. Seroprevalence is impacted by multiple factors, including geography, age, capsid serotype, and assay type. Anti-AAV antibody assays typically measure (1) transduction inhibition by detecting the neutralizing capacity of antibodies and non-antibody neutralizing factors, or (2) total anti-capsid binding antibodies, regardless of neutralizing activity. Presently, there is a paucity of head-to-head data and standardized approaches associating assay results with clinical outcomes. In addition, establishing clinically relevant screening titer cutoffs is complex. Thus, meaningful comparisons across assays are nearly impossible. Although complex, establishing screening assays in routine clinical practice to identify patients with antibody levels that may impact favorable treatment outcomes is achievable for both transduction inhibition and total antibody assays. Formal regulatory approval of such assays as companion diagnostic tests will confirm their suitability for specific recombinant AAV gene therapies. This review covers current approaches to measure anti-AAV antibodies in patient plasma or serum, their potential impact on therapeutic safety and efficacy, and investigative strategies to mitigate the effects of pre-existing anti-AAV antibodies in patients.
Collapse
Affiliation(s)
- Martin Schulz
- Pfizer, 235 East 42nd Street, New York, NY 10017, USA
| | - Daniel I Levy
- Pfizer, 235 East 42nd Street, New York, NY 10017, USA
| | | | | | - Ian Winburn
- Pfizer, 235 East 42nd Street, New York, NY 10017, USA
| | - Matthias Mahn
- Pfizer, 235 East 42nd Street, New York, NY 10017, USA
| | | | - Seng H Cheng
- Pfizer, 235 East 42nd Street, New York, NY 10017, USA
| | - Barry J Byrne
- University of Florida, 1600 SW Archer Road, Gainesville, FL 32610, USA.
| |
Collapse
|
24
|
Breast milk from healthy women has higher anti- Candida properties than women with vaginal infections during pregnancy. Food Sci Biotechnol 2023; 32:471-480. [PMID: 36911325 PMCID: PMC9992674 DOI: 10.1007/s10068-022-01088-x] [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: 03/15/2022] [Revised: 04/07/2022] [Accepted: 04/10/2022] [Indexed: 11/04/2022] Open
Abstract
The aim of this study was to investigate the different immunological and antimicrobial properties of breast milk from women with (W) or without (WO) vaginal yeast infections during pregnancy in 85 lactating women (W, n = 43; WO, n = 42). Concentrations of IL-10, IgA, IgM, IgG, EGF, and TGF-α were similar in both groups. However, breast milk of women aged below 31 years old from the W-group showed higher concentration of EGF than the WO-group (p = 0.031). Breast milk from WO-group exhibited higher anti-Candida properties than W-group, both via growth inhibition and aggregation of yeast cells (p < 0.001). Correlation analysis showed that breast milk concentration of TGF-α positively correlated with concentrations of IL-10 (p = 0.001) and IgA (p = 0.021) in the W-group. Data from our present study shows that although breast milk from women with vaginal infections during pregnancy may not sufficiently hinder Candida growth, other immuno-modulatory bioactives may substitute for such a protective effect.
Collapse
|
25
|
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: 2.0] [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
|
26
|
Bigler NA, Bruckmaier RM, Gross JJ. Implications of placentation type on species-specific colostrum properties in mammals. J Anim Sci 2022; 100:skac287. [PMID: 36048628 PMCID: PMC9713508 DOI: 10.1093/jas/skac287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/30/2022] [Indexed: 11/12/2022] Open
Abstract
Maternal care is essential to optimally support survival of the offspring. During evolution of mammalian species, different phenotypes have evolved in relation to gestation length, number, size, and maturation stage of the offspring at parturition, as well as colostrum and milk composition. The aim of the present review is to describe relationships between placental function and colostrum and milk composition in different mammalian species. Species covered in this article include humans, rabbits, rodents (rat and mouse), carnivores (cats and dogs), and a variety of ungulate species (cattle, sheep, goats, pigs, and horses). Species-specific aspects are elucidated with a special focus on the transfer of passive immunity. In this regard, the structure and thus the capability of the placenta to transport immunoglobulins from maternal to fetal circulation in utero dictates the necessity of the passive transfer of immunity via colostrum. Consequently, species with exclusive postpartal transfer of immunity such as in all ungulate species have greater immunoglobulin G concentrations in colostrum than species with a prepartal transfer in utero, where especially immunoglobulin A with its local immune function in the gastrointestinal tract is present in colostrum (e.g., rabbit and human). In terms of the nutritional purpose, suckling frequency is an important factor determining the gross composition of colostrum as well as in the mature milk of these species. Milk of nidicolous animals with long intervals in-between suckling events contains more fat than milk of nidifugous animals with constant access to their mother. However, the importance of colostrum and milk consumption for newborn animals and human babies goes beyond nutrition and the transfer of immunity. Numerous bioactive components such as growth factors, hormones, and oligosaccharides are enriched in colostrum and transition milk, which support the development of the intestinal tract and local immune system.
Collapse
Affiliation(s)
- Naomi A Bigler
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Rupert M Bruckmaier
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Josef J Gross
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
27
|
Manoppo JIC, Nurkolis F, Gunawan WB, Limen GA, Rompies R, Heroanto JP, Natanael H, Phan S, Tanjaya K. Functional sterol improves breast milk quality by modulating the gut microbiota: A proposed opinion for breastfeeding mothers. Front Nutr 2022; 9:1018153. [PMID: 36424924 PMCID: PMC9678907 DOI: 10.3389/fnut.2022.1018153] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/16/2022] [Indexed: 09/30/2023] Open
Affiliation(s)
- Jeanette Irene Christiene Manoppo
- Department of Pediatrics, Faculty of Medicine, Sam Ratulangi University, Manado, Indonesia
- Department of Pediatrics, Prof. R. D. Kandou General Hospital, Manado, Indonesia
| | - Fahrul Nurkolis
- Biological Sciences, Faculty of Sciences and Technology, State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga Yogyakarta), Yogyakarta, Indonesia
| | - William Ben Gunawan
- Department of Nutrition Science, Faculty of Medicine, Diponegoro University, Semarang, Indonesia
| | - Gilbert Ansell Limen
- Medical Programme, Faculty of Medicine, Sam Ratulangi University, Manado, Indonesia
| | - Ronald Rompies
- Department of Pediatrics, Faculty of Medicine, Sam Ratulangi University, Manado, Indonesia
- Department of Pediatrics, Prof. R. D. Kandou General Hospital, Manado, Indonesia
| | - Joko Purnomo Heroanto
- Department of Pediatrics, Faculty of Medicine, Sam Ratulangi University, Manado, Indonesia
- Department of Pediatrics, Prof. R. D. Kandou General Hospital, Manado, Indonesia
| | - Hans Natanael
- Department of Pediatrics, Faculty of Medicine, Sam Ratulangi University, Manado, Indonesia
- Department of Pediatrics, Prof. R. D. Kandou General Hospital, Manado, Indonesia
| | - Sardito Phan
- Department of Pediatrics, Faculty of Medicine, Sam Ratulangi University, Manado, Indonesia
- Department of Pediatrics, Prof. R. D. Kandou General Hospital, Manado, Indonesia
| | - Krisanto Tanjaya
- Medical Programme, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| |
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: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 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
|
Longueira Y, Ojeda DS, Battistelli RBA, Sanchez L, Oviedo Rouco S, Albano D, Guevara E, Valls V, Pando MA, Gamarnik AV. SARS-CoV-2-Specific IgG and IgA response in maternal blood and breastmilk of vaccinated naïve and convalescent lactating participants. Front Immunol 2022; 13:909995. [PMID: 36263055 PMCID: PMC9574440 DOI: 10.3389/fimmu.2022.909995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 09/01/2022] [Indexed: 12/31/2022] Open
Abstract
Background Recent studies have shown the presence of SARS-CoV-2-specific antibodies in the milk of breastfeeding mothers vaccinated with mRNA and convalescent. However, limited information is available in lactating women receiving other vaccine platforms used in developing countries, such as the inactivated SARS-CoV-2 vaccine BBIBP-CorV (Sinopharm) and the non-replicating adenovirus vaccines Sputnik V (Gamaleya Institute) and ChAdOx1-S (Oxford AstraZeneca). Methods Here, we evaluated anti-SARS-CoV-2 IgG and IgA levels in both serum and milk samples using a longitudinal and a cross-sectional cohort of 208 breastfeeding vaccinated women from Argentina with or without previous SARS-CoV-2 infection. Results The analysis showed that IgA levels remain constant in serum and milk of breastfeeding mothers between the first and second doses of vector-based vaccines (Sputnik V and ChAdOx1-S). After the second dose, anti-spike IgA was found positive in 100% of the serum samples and in 66% of breastmilk samples. In addition, no significant differences in milk IgA levels were observed in participants receiving BBIBP-CorV, Sputnik V or ChAdOx1-S. IgG levels in milk increased after the second dose of vector-based vaccines. Paired longitudinal samples taken at 45 and 120 days after the second dose showed a decrease in milk IgG levels over time. Study of IgA levels in serum and milk of vaccinated naïve of infection and vaccinated-convalescent breastfeeding participants showed significantly higher levels in vaccinated-convalescent than in participants without previous infection. Conclusion This study is relevant to understand the protection against SARS-CoV-2 by passive immunity in newborns and children who are not yet eligible to receive vaccination.
Collapse
Affiliation(s)
- Yesica Longueira
- CONICET – Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina
| | - Diego S. Ojeda
- Laboratorio de Virología Molecular, Fundación Instituto Leloir-CONICET, Buenos Aires, Argentina,Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología, Buenos Aires, Argentina,*Correspondence: Andrea V. Gamarnik, ; Diego S. Ojeda,
| | - Rocio B. Antivero Battistelli
- CONICET – Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina,Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología, Buenos Aires, Argentina
| | - Lautaro Sanchez
- Laboratorio de Virología Molecular, Fundación Instituto Leloir-CONICET, Buenos Aires, Argentina
| | - Santiago Oviedo Rouco
- Laboratorio de Virología Molecular, Fundación Instituto Leloir-CONICET, Buenos Aires, Argentina
| | - Daniel Albano
- Banco de Leche Humana – Hospital Materno Infantil Ramón Sardá, Ciudad Autónoma de Buenos Aires, Argentina
| | - Eleonora Guevara
- Banco de Leche Humana – Hospital Materno Infantil Ramón Sardá, Ciudad Autónoma de Buenos Aires, Argentina
| | - Vanesa Valls
- Banco de Leche Humana – Hospital Materno Infantil Ramón Sardá, Ciudad Autónoma de Buenos Aires, Argentina
| | - María A. Pando
- CONICET – Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina,Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología, Buenos Aires, Argentina
| | - Andrea V. Gamarnik
- Laboratorio de Virología Molecular, Fundación Instituto Leloir-CONICET, Buenos Aires, Argentina,*Correspondence: Andrea V. Gamarnik, ; Diego S. Ojeda,
| |
Collapse
|
30
|
Liang N, Koh J, Kim BJ, Ozturk G, Barile D, Dallas DC. Structural and functional changes of bioactive proteins in donor human milk treated by vat-pasteurization, retort sterilization, ultra-high-temperature sterilization, freeze-thawing and homogenization. Front Nutr 2022; 9:926814. [PMID: 36185694 PMCID: PMC9521613 DOI: 10.3389/fnut.2022.926814] [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: 04/23/2022] [Accepted: 08/22/2022] [Indexed: 11/26/2022] Open
Abstract
Background Donor human milk should be processed to guarantee microbiological safety prior to infant feeding, but this process can influence the structure and quantity of functional proteins. Objective The aim of this study was to determine the effect of thawing, homogenization, vat-pasteurization (Vat-PT), retort sterilization (RTR) and ultra-high-temperature (UHT) processing on the structure of bioactive proteins in donor milk. Methods Pooled donor milk was either not treated (Raw) or treated with an additional freeze-thaw cycle with and without homogenization, Vat-PT, RTR with and without homogenization, and UHT processing with and without homogenization. Overall protein retention was assessed via sodium-dodecyl sulfate (SDS-PAGE), and the immunoreactivity of 13 bioactive proteins were assessed via enzyme-linked immunosorbent assay (ELISA). Results Freeze-thawing, freeze-thawing plus homogenization and Vat-PT preserved all the immunoglobulins (sIgA/IgA, IgG, IgM) in donor milk, whereas RTR and UHT degraded almost all immunoglobulins. UHT did not alter osteopontin immunoreactivity, but Vat-PT and retort decreased it by ~50 and 70%, respectively. Freeze-thawing with homogenization, Vat-PT and UHT reduced lactoferrin's immunoreactivity by 35, 65, and 84%, respectively. Lysozyme survived unaltered throughout all processing conditions. In contrast, elastase immunoreactivity was decreased by all methods except freeze-thawing. Freeze-thawing, freeze-thawing plus homogenization and Vat-PT did not alter polymeric immunoglobulin receptor (PIGR) immunoreactivity, but RTR, RTR plus homogenization and UHT increased detection. All heat processing methods increased α-lactalbumin immunoreactivity. Vat-PT preserved all the growth factors (vascular/endothelial growth factor, and transforming growth factors β1 and β2), and UHT treatments preserved the majority of these factors. Conclusion Different bioactive proteins have different sensitivity to the treatments tested. Overall, Vat-PT preserved more of the bioactive proteins compared with UHT or RTR. Therefore, human milk processors should consider the impact of processing methods on key bioactive proteins in human milk.
Collapse
Affiliation(s)
- Ningjian Liang
- Nutrition Program, School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, United States
| | - Jeewon Koh
- Nutrition Program, School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, United States
| | - Bum Jin Kim
- Nutrition Program, School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, United States
| | - Gulustan Ozturk
- Department of Food Science and Technology, University of California, Davis, Davis, CA, United States
| | - Daniela Barile
- Department of Food Science and Technology, University of California, Davis, Davis, CA, United States
| | - David C. Dallas
- Nutrition Program, School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, United States
| |
Collapse
|
31
|
Huang YJ, Porsche C, Kozik AJ, Lynch SV. Microbiome-Immune Interactions in Allergy and Asthma. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:2244-2251. [PMID: 35724951 PMCID: PMC10566566 DOI: 10.1016/j.jaip.2022.05.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/13/2022] [Accepted: 05/28/2022] [Indexed: 06/13/2023]
Abstract
The human microbiota has been established as a key regulator of host health, in large part owing to its constant interaction with and impact on host immunity. A range of environmental exposures spanning from the prenatal period through adulthood are known to affect the composition and molecular productivity of microbiomes across mucosal and dermal tissues with short- and long-term consequences for host immune function. Here we review recent findings in the field that provide insights into how microbial-immune interactions promote and sustain immune dysfunction associated with allergy and asthma. We consider both early life microbiome perturbation and the molecular underpinnings of immune dysfunction associated with subsequent allergy and asthma development in childhood, as well as microbiome features that relate to phenotypic attributes of allergy and asthma in older patients with established disease.
Collapse
Affiliation(s)
- Yvonne J Huang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan, Ann Arbor, Mich; Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Mich.
| | - Cara Porsche
- Department of Medicine, University of California San Francisco, San Francisco, Calif
| | - Ariangela J Kozik
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan, Ann Arbor, Mich
| | - Susan V Lynch
- Department of Medicine, University of California San Francisco, San Francisco, Calif.
| |
Collapse
|
32
|
Ricciardi A, Zelini P, Cassaniti I, Avanzini MA, Colaneri M, De Silvestri A, Baldanti F, Bruno R. Serum and breastmilk SARS-CoV-2 specific antibodies following BNT162b2 vaccine: prolonged protection from SARS-CoV-2 in newborns and older children. Int J Infect Dis 2022; 122:905-909. [PMID: 35803470 PMCID: PMC9254442 DOI: 10.1016/j.ijid.2022.06.055] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVES Vaccination is the best strategy against COVID-19. We aimed to determine antibodies against SARS-CoV-2 in breastmilk and serum of mothers vaccinated with the mRNA vaccine. METHODS This prospective study included 18 lactating women vaccinated with the BNT162b2 vaccine. Serum and breastmilk were collected before the first dose (T0), at the second dose (T1), 3 weeks after the second dose (T2), and 6 months after the first dose (T3). Serum anti-SARS-CoV-2 Spike (S) Immunoglobulin G (IgG) and Immunoglobulin A (IgA) were measured using a semi-quantitative enzyme-linked immunosorbent assay (ELISA) and secretory antibody (s) IgG and IgA in breastmilk using quantitative analysis. RESULTS We detected serum anti-S IgG and IgA in all women after vaccination. Specific IgG and IgA were higher at T1, T2, and T3 compared with T0 (P <0.0001). Higher antibody levels were observed at T2 and lower values at T3 versus T2 (P = 0.007). After 6 months, all patients had serum IgG, but three of 18 (16%) had serum IgA. In breastmilk, sIgA was present at T1 and T2 and decreased after 6 months at T3 (P = 0.002). Breastmilk sIgG levels increased at T1 and T2 and peaked at T3 (P = 0.008). CONCLUSION Secretory antibodies were transmitted through breastmilk until 6 months after anti-COVID-19 mRNA vaccination. Protection of the newborn through breastfeeding needs to be addressed.
Collapse
Affiliation(s)
- Alessandra Ricciardi
- Department of Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| | - Paola Zelini
- Department of Obstetrics and Gynecology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Irene Cassaniti
- Molecular Virology Unit, Department of Microbiology and Virology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Maria Antonietta Avanzini
- Cell Factory, Pediatric Hematology Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marta Colaneri
- Department of Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Annalisa De Silvestri
- Service of Biometry and Statistics Fondazione IRCCS Policlinico San Matteo, Viale Camillo Golgi 19, 27100, Pavia, Italy
| | - Fausto Baldanti
- Molecular Virology Unit, Department of Microbiology and Virology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Raffaele Bruno
- Department of Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
33
|
Exploring the Potential of Human Milk and Formula Milk on Infants’ Gut and Health. Nutrients 2022; 14:nu14173554. [PMID: 36079814 PMCID: PMC9460722 DOI: 10.3390/nu14173554] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/21/2022] Open
Abstract
Early-life gut microbiota plays a role in determining the health and risk of developing diseases in later life. Various perinatal factors have been shown to contribute to the development and establishment of infant gut microbiota. One of the important factors influencing the infant gut microbial colonization and composition is the mode of infant feeding. While infant formula milk has been designed to resemble human milk as much as possible, the gut microbiome of infants who receive formula milk differs from that of infants who are fed human milk. A diverse microbial population in human milk and the microbes seed the infant gut microbiome. Human milk contains nutritional components that promote infant growth and bioactive components, such as human milk oligosaccharides, lactoferrin, and immunoglobulins, which contribute to immunological development. In an attempt to encourage the formation of a healthy gut microbiome comparable to that of a breastfed infant, manufacturers often supplement infant formula with prebiotics or probiotics, which are known to have a bifidogenic effect and can modulate the immune system. This review aims to elucidate the roles of human milk and formula milk on infants’ gut and health.
Collapse
|
34
|
Pieri M, Maniori MA, Shahabian L, Kanaan E, Paphiti-Demetriou I, Pipis S, Felekkis K, Nicolaidou V, Papaneophytou C. Survival of Vaccine-Induced Human Milk SARS-CoV-2 IgG, IgA and SIgA Immunoglobulins across Simulated Human Infant Gastrointestinal Digestion. Nutrients 2022; 14:3368. [PMID: 36014873 PMCID: PMC9415426 DOI: 10.3390/nu14163368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
Breastfeeding can be a vital way of acquiring passive immunity via the transfer of antibodies from the mother to the breastfeeding infant. Recent evidence points to the fact that human milk contains immunoglobulins (Ig) against the SARS-CoV-2 virus, either after natural infection or vaccination, but whether these antibodies can resist enzymatic degradation during digestion in the infant gastrointestinal (GI) tract or indeed protect the consumers remains inconclusive. Herein, we evaluated the levels of IgG, IgA, and secretory IgA (SIgA) antibodies against the spike protein of SARS-CoV-2 in 43 lactating mothers who received at least two doses of either an mRNA-based vaccine (Pfizer/BioNTech, Moderna; n = 34) or an adenovirus-based vaccine (AstraZeneca; n = 9). We also accessed the potential persistence of SARS-CoV-2 IgA, IgG, and secretory IgA (SIgA) antibodies from vaccinated women in the GI tract of the infants by means of a static in vitro digestion protocol. Our data depict that, although slightly reduced, the IgA antibodies produced after vaccination resist both the gastric and intestinal phases of infant digestion, whereas the IgGs are more prone to degradation in both phases of digestion. Additionally, SIgA antibodies were found to greatly resist the gastric phase of digestion albeit showing some reduction during the intestinal phase. The evaluation of the vaccine induced Ig profile of breastmilk, and the extent to which these antibodies can resist digestion in the infant GI tract provide important information about the potential protective role of this form of passive immunity that could help decision making during the COVID-19 pandemic and beyond.
Collapse
Affiliation(s)
- Myrtani Pieri
- Department of Life and Health Sciences, University of Nicosia, 46 Makedonitissas Avenue, Nicosia CY-1700, Cyprus
| | - Maria-Arsenia Maniori
- Department of Life and Health Sciences, University of Nicosia, 46 Makedonitissas Avenue, Nicosia CY-1700, Cyprus
| | - Lucy Shahabian
- Department of Life and Health Sciences, University of Nicosia, 46 Makedonitissas Avenue, Nicosia CY-1700, Cyprus
| | - Elie Kanaan
- Department of Life and Health Sciences, University of Nicosia, 46 Makedonitissas Avenue, Nicosia CY-1700, Cyprus
| | | | - Spyros Pipis
- Medical School, University of Nicosia, Nicosia CY-2408, Cyprus
- Aretaeio Hospital, 55-57, Andrea Avraamidi St, Strovolos, Nicosia CY-2024, Cyprus
| | - Kyriakos Felekkis
- Department of Life and Health Sciences, University of Nicosia, 46 Makedonitissas Avenue, Nicosia CY-1700, Cyprus
| | - Vicky Nicolaidou
- Department of Life and Health Sciences, University of Nicosia, 46 Makedonitissas Avenue, Nicosia CY-1700, Cyprus
| | - Christos Papaneophytou
- Department of Life and Health Sciences, University of Nicosia, 46 Makedonitissas Avenue, Nicosia CY-1700, Cyprus
| |
Collapse
|
35
|
Warner SA, Arevalo JL. Literature Review of Mothers Diagnosed With COVID-19 and the Impact on Breastfeeding Their Newborns. Nurs Womens Health 2022; 26:234-241. [PMID: 35551888 PMCID: PMC9611848 DOI: 10.1016/j.nwh.2022.03.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 03/04/2022] [Accepted: 03/29/2022] [Indexed: 01/09/2023]
Abstract
In early 2020, newborns were separated from their mothers who were diagnosed with COVID-19 infection. The purpose of this literature review is to examine the current evidence to determine if the transmission of COVID-19 infection to the neonate increases if newborns are directly breastfed by mothers who are positive for COVID-19 infection. There were 28 studies conducted in seven countries, with 10 of those studies conducted in the United States. In total, 5,123 neonates were born to mothers diagnosed with COVID-19, with 3,872 neonates determined to have been breastfed or provided mixed feeding including breast milk. Overall, 2.35% (91/3,872) of the newborns tested positive, and all recovered from COVID-19 infection. As frontline health care providers, nurses are instrumental in offering support and education on the risks and benefits of breastfeeding for individuals diagnosed with COVID-19.
Collapse
|
36
|
Esteve-Palau E, Gonzalez-Cuevas A, Eugenia Guerrero M, Garcia-Terol C, Carmen Alvarez M, Garcia G, Moreno E, Medina F, Casadevall D, Diaz-Brito V. Quantification and progress over time of specific antibodies against SARS-CoV-2 in breast milk of lactating women vaccinated with BNT162b2 Pfizer-BioNTech COVID-19 vaccine (LacCOVID). Open Forum Infect Dis 2022; 9:ofac239. [PMID: 35783685 PMCID: PMC9129184 DOI: 10.1093/ofid/ofac239] [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: 01/03/2022] [Accepted: 05/09/2022] [Indexed: 11/29/2022] Open
Abstract
Background Several observational studies demonstrated the passage of postvaccine antibodies through breast milk in women vaccinated against coronavirus disease 2019 (COVID-19), mostly with messenger RNA (mRNA)–based vaccines, but lacked long-term data. Methods A 6-month prospective cohort study was performed to determine severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine–induced antibody levels in the breast milk of 33 lactating healthcare workers at different timepoints after mRNA BNT162b2 Pfizer-BioNTech COVID-19 vaccination. Moreover, we examined the correlation of SARS-CoV-2 antibody levels between serum and breast milk, adverse events related to vaccination, and rate of SARS-CoV-2 infections. Results Mothers’ median age was 38 (interquartile range [IQR], 36–39) years and 15 (IQR, 10–22) months for infants. Median (IQR) SARS-CoV-2 immunoglobulin G (IgG) spike protein subunit S1 (S1) vaccine–induced levels at different timepoints for serum–milk pairs were 519 (234–937) to 1 (0–2.9) arbitrary units (AU)/mL at 2 weeks after first dose and 18 644 (9923–29 264) to 78 (33.7–128), 12 478 (6870–20 801) to 50.4 (24.3–104), 4094 (2413–8480) to 19.9 (10.8–51.9), 1350 (831–2298) to 8.9 (7.8–31.5) AU/mL at 2, 4, 12 and 24 weeks after second dose, respectively. We observed a positive correlation of antibody levels between serum and breast milk, no serious adverse events related to vaccination, and 2 (6%) COVID-19 vaccine breakthrough infections. Conclusions Women vaccinated with Pfizer-BioNTech transmit antibodies into breast milk with a positive correlation with serum levels. Both decreased over time in a 6-month follow-up.
Collapse
Affiliation(s)
- Erika Esteve-Palau
- Department of Infectious Diseases, Parc Sanitari Sant Joan de Deu (Sant Boi, Barcelona, Spain)
| | | | - M. Eugenia Guerrero
- Department of Microbiology, Parc Sanitari Sant Joan de Deu (Sant Boi, Barcelona, Spain)
| | - Clara Garcia-Terol
- Department of Obstetrics and Gynecology, Parc Sanitari Sant Joan de Deu (Sant Boi, Barcelona, Spain)
| | - M. Carmen Alvarez
- Department of Infectious Diseases, Parc Sanitari Sant Joan de Deu (Sant Boi, Barcelona, Spain)
| | - Geneva Garcia
- Department of Infectious Diseases, Parc Sanitari Sant Joan de Deu (Sant Boi, Barcelona, Spain)
| | - Encarna Moreno
- Department of Infectious Diseases, Parc Sanitari Sant Joan de Deu (Sant Boi, Barcelona, Spain)
| | - Francisco Medina
- Department of Infectious Diseases, Parc Sanitari Sant Joan de Deu (Sant Boi, Barcelona, Spain)
| | - David Casadevall
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Vicens Diaz-Brito
- Department of Infectious Diseases, Parc Sanitari Sant Joan de Deu (Sant Boi, Barcelona, Spain)
| |
Collapse
|
37
|
Langel SN, Blasi M, Permar SR. Maternal immune protection against infectious diseases. Cell Host Microbe 2022; 30:660-674. [PMID: 35550669 DOI: 10.1016/j.chom.2022.04.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The maternal immune system protects developing offspring against pathogens before birth via transplacental transfer and after birth through secreted milk. This transferred maternal immunity influences each generation's susceptibility to infections and responsiveness to immunization. Thus, boosting immunity in the maternal-neonatal dyad is a potentially valuable public health strategy. Additionally, at critical times during fetal and postnatal development, environmental factors and immune stimuli influence immune development. These "windows of opportunity" offer a chance to identify both risk and protective factors that promote long-term health and limit disease. Here, we review pre- and postpartum maternal immune factors that protect against infectious agents in offspring and how they may shape the infant's immune landscape over time. Additionally, we discuss the influence of maternal immunity on the responsiveness to immunization in early life. Lastly, when maternal factors are insufficient to prevent neonatal infectious diseases, we discuss pre- and postnatal therapeutic strategies for the maternal-neonatal dyad.
Collapse
Affiliation(s)
- Stephanie N Langel
- Department of Surgery, Duke Center for Human Systems Immunology, Durham, NC, USA
| | - Maria Blasi
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA; Department of Medicine, Division of Infectious Diseases, Duke University Medical Center, Durham, NC, USA
| | - Sallie R Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
38
|
Juncker HG, Mulleners SJ, Ruhé EJ, Coenen ER, Bakker S, van Doesburg M, Harinck JE, Rood RD, Bouhuijs JH, Oomen M, de Groot PCJ, Pajkrt PD, Korosi A, van Goudoever PJB, van Gils MJ, van Keulen BJ. Comparing the human milk antibody response after vaccination with four COVID-19 vaccines: A prospective, longitudinal cohort study in the Netherlands. EClinicalMedicine 2022; 47:101393. [PMID: 35465077 PMCID: PMC9013951 DOI: 10.1016/j.eclinm.2022.101393] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/22/2022] [Accepted: 03/28/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Vaccination of lactating women against COVID-19 may protect not only themselves but also their breastfed infant through human milk. Therefore, it is important to gain insight into the human milk antibody response after immunization with the various vaccines that are currently widely used. The aim of this study is to determine and compare the antibody response in human milk following vaccination with mRNA- and vector-based vaccines up to over two months post-vaccination. METHODS This prospective cohort study was conducted in the Netherlands between January 06, 2021 and July 31, 2021. Participants were recruited through social media. Human milk samples were collected longitudinally during a period of 70 days from women receiving one of the four different severe acute respiratory coronavirus 2 (SARS-CoV-2) vaccines: Pfizer-BioNTech (BNT162b2), Moderna (mRNA-1273), Oxford/AstraZeneca (AZD1222) and Johnson&Johnson (Ad26.COV2.S). SARS-CoV-2-specific antibodies were measured using an enzyme-linked immunosorbent assay. The area under the curve (AUC) of the Immunoglobulins A (IgA) and G (IgG) antibody response was determined over 15 and 70 days following the first vaccination and compared between the different vaccines. FINDINGS This study enrolled 134 vaccinated lactating women of whom 97 participated the entire study period. In total, 1887 human milk samples were provided. The human milk antibody response differed between SARS-CoV-2 vaccines over the study period. The mean AUC of SARS-CoV-2-specific IgA, but not IgG, in human milk over 15 days was higher after vaccination with an mRNA-based vaccine than a vector-based vaccine (AUC with respect to ground [AUCg] ± the standard error of the mean [SEM] for IgA was 6·09 ± 0·89 in the BNT162b2 group, 7·48 ± 1·03 in the mRNA-1273 group, 4·17 ± 0·73 in the AZD1222 group, and 5·71 ± 0·70 in the Ad26.COV2.S group). Over a period of 70 days, the mean AUCg of both IgA and IgG was higher after vaccination with an mRNA-based vaccine than a vector-based vaccine (AUCg ± SEM for IgA was 38·77 ± 6·51 in the BNT162b2 group, 50·13 ± 7·41 in the mRNA-1273 group, 24·12 ± 5·47 in the AZD1222 group, and 28·15 ± 6·69 in the Ad26.COV2.S group; AUCg ± SEM for IgG was 40·43 ± 2·67 in the BNT162b2 group, 37·01 ± 2·38 in the mRNA-1273 group, 16·04 ± 5·09 in the AZD1222 group, and 10·44 ± 2·50 in the Ad26.COV2.S group). INTERPRETATION Overall, maternal vaccination during lactation with an mRNA-based vaccine resulted in higher SARS-CoV-2 antibody responses in human milk compared to vector-based vaccines. Therefore, vaccination with mRNA-based vaccines, preferably with the mRNA-1273 vaccine, might not only provide better immunological protection for the mother but also for her breastfed infant. FUNDING Stichting Steun Emma Kinderziekenhuis and the Amsterdam Infection and Immunity Institute (grant 24175).
Collapse
Affiliation(s)
- Hannah G. Juncker
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
- Swammerdam Institute for Life Sciences - Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - Sien J. Mulleners
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
| | - Eliza J.M. Ruhé
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
| | - Esmée R.M. Coenen
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
| | - Sjors Bakker
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
| | - Maritt van Doesburg
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
| | - Jolinda E. Harinck
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
| | - Romee D. Rood
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
| | - Joey H. Bouhuijs
- Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Department of Medical Microbiology and Infection Prevention, Amsterdam, the Netherlands
| | - Melissa Oomen
- Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Department of Medical Microbiology and Infection Prevention, Amsterdam, the Netherlands
| | - Prof. Christianne J.M. de Groot
- Amsterdam UMC, Vrije Universiteit, Amsterdam Reproduction & Development Research Institute, Department of Obstetrics and Gynaecology, Amsterdam, the Netherlands
| | - Prof. Dasja Pajkrt
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
| | - Aniko Korosi
- Swammerdam Institute for Life Sciences - Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - Prof. Johannes B. van Goudoever
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
- Corresponding author at: Hans van Goudoever, Emma Children's Hospital - Amsterdam UMC, Meibergdreef 9, 1000 DE Amsterdam, The Netherlands. tel +31-20-5668885.
| | - Marit J. van Gils
- Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Department of Medical Microbiology and Infection Prevention, Amsterdam, the Netherlands
| | - Britt J. van Keulen
- Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam Reproduction & Development Research Institute, Department of Pediatrics, Amsterdam, the Netherlands
| |
Collapse
|
39
|
Trofin F, Nastase EV, Iancu LS, Constantinescu D, Cianga CM, Lunca C, Ursu RG, Cianga P, Dorneanu OS. Anti-RBD IgA and IgG Response and Transmission in Breast Milk of Anti-SARS-CoV-2 Vaccinated Mothers. Pathogens 2022; 11:286. [PMID: 35335610 PMCID: PMC8952534 DOI: 10.3390/pathogens11030286] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 12/24/2022] Open
Abstract
The appearance of the severe acute respiratory syndrome virus-2 (SARS-CoV-2) has had a significant impact on the balance of public health and social life. The data available so far show that newborns and young children do not develop severe forms of COVID-19, but a small proportion of them will still need hospitalization. Even though young children represent an important vector of the infection, vaccination at such a young age was not yet considered. Thus, the question of whether potentially protective antibodies against SARS-CoV-2 could be provided to them via breast milk or across the placenta, as "passive immunity", still stands. MATERIALS AND METHODS Between January-July 2021, we have conducted a prospective study that aimed to measure the immunoglobulin (Ig) A and IgG anti-SARS-CoV-2 titers in the breast milk of 28 vaccinated lactating mothers, sampled at 30 and 60 days after the second dose of the anti-SARS-CoV-2 Pfizer or Moderna mRNA vaccines. Anti-RBD reactive IgA and IgG antibodies were detected and quantified by a sandwich enzyme-linked immunosorbent assay. RESULTS Anti-RBD IgA and IgG were present in all breast milk samples, both in the first and in the second specimens, without a significant difference between those two. The anti-RBD IgA titers were approximately five-times higher than the anti-RBD IgG ones. The anti-RBD IgA and IgG titers were correlated with the infants' age, but they were not correlated with the vaccine type or mother's age. The anti-RBD IgA excreted in milk were inversely correlated with the parity number. CONCLUSIONS Anti-SARS-CoV-2 IgA and IgG can be found in the milk secretion of mothers vaccinated with mRNA vaccines and, presumably, these antibodies should offer protection to the newborn, considering that the antibodies' titers did not decrease after 60 days. The antibody response is directly proportional to the breastfed child's age, but the amount of anti-RBD IgA decreases with the baby's rank. The antibody response did not depend on the vaccine type, or on the mother's age.
Collapse
Affiliation(s)
- Felicia Trofin
- Microbiology Department, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (F.T.); (C.L.); (R.G.U.); (O.S.D.)
- Clinical Hospital of Infectious Diseases “Sf. Parascheva”, 700116 Iasi, Romania;
| | - Eduard Vasile Nastase
- Clinical Hospital of Infectious Diseases “Sf. Parascheva”, 700116 Iasi, Romania;
- Infectious Diseases Department, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| | - Luminita Smaranda Iancu
- Microbiology Department, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (F.T.); (C.L.); (R.G.U.); (O.S.D.)
- National Institute of Public Health, Iasi Regional Center for Public Health, 700465 Iasi, Romania
| | - Daniela Constantinescu
- Immunology Department, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (D.C.); (C.M.C.)
- Immunology Laboratory, “Sf. Spiridon” Clinical Hospital, 700111 Iasi, Romania
| | - Corina Maria Cianga
- Immunology Department, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (D.C.); (C.M.C.)
- Immunology Laboratory, “Sf. Spiridon” Clinical Hospital, 700111 Iasi, Romania
| | - Catalina Lunca
- Microbiology Department, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (F.T.); (C.L.); (R.G.U.); (O.S.D.)
- National Institute of Public Health, Iasi Regional Center for Public Health, 700465 Iasi, Romania
| | - Ramona Gabriela Ursu
- Microbiology Department, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (F.T.); (C.L.); (R.G.U.); (O.S.D.)
| | - Petru Cianga
- Immunology Department, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (D.C.); (C.M.C.)
- Immunology Laboratory, “Sf. Spiridon” Clinical Hospital, 700111 Iasi, Romania
| | - Olivia Simona Dorneanu
- Microbiology Department, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (F.T.); (C.L.); (R.G.U.); (O.S.D.)
- Clinical Hospital of Infectious Diseases “Sf. Parascheva”, 700116 Iasi, Romania;
| |
Collapse
|
40
|
Scrimin F, Campisciano G, Comar M, Ragazzon C, Davanzo R, Quadrifoglio M, Giangreco M, Stabile G, Ricci G. IgG and IgA Antibodies Post SARS-CoV-2 Vaccine in the Breast Milk and Sera of Breastfeeding Women. Vaccines (Basel) 2022; 10:vaccines10010125. [PMID: 35062786 PMCID: PMC8778843 DOI: 10.3390/vaccines10010125] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 02/04/2023] Open
Abstract
The COVID-19 pandemic has carried massive global health and economic burden that is currently counteracted by a challenging anti-COVID-19 vaccination campaign. Indeed, mass vaccination against COVID-19 is expected to be the most efficacious intervention to mitigate the pandemic successfully. The primary objective of the present study is to test the presence of neutralizing anti-SARS-CoV-2 antibodies (IgA and IgG) in the breast milk and sera samples from vaccinated women at least 20 days after the complete vaccine cycle. A secondary aim is to compare the IgG antibodies level in maternal serum and breast milk. The third target is to evaluate the presence of the IgG antibodies in breast milk after several weeks from the vaccination. Finally, we collected information on the health status of infants in the days following maternal vaccination. Forty-two mothers were enrolled in the study. Thirty-six received the Pfizer/BioNTech vaccine, four the Astra Zeneca vaccine, one the Moderna vaccine and another woman Astra Zeneca in the first dose and Pfizer/BioNTech in the second dose. All 42 milk samples confirmed the presence of anti-SARS-CoV-2 IgG, and none showed IgA presence. Regarding the matched 42 sera samples, 41 samples detected IgG presence, with one sample testing negative and only one positive for seric IgA. None of the 42 infants had fever or changes in sleep or appetite in the seven days following the maternal vaccination. The level of IgG antibodies in milk was, on average, lower than that in maternal serum. According to our analysis, the absence of IgA could suggest a rapid decrease after vaccination even if frequent breastfeeding could favour its persistence. IgG were present in breast milk even 4 months after the second vaccine dose. Information on the immunological characteristics of breast milk could change mothers’ choices regarding breastfeeding.
Collapse
Affiliation(s)
- Federica Scrimin
- Department of Obstetrics and Gynaecology, Institute for Maternal and Child Health-IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (F.S.); (M.Q.); (G.R.)
| | - Giuseppina Campisciano
- Department of Advanced Microbiology Diagnosis and Translational Research, Institute for Maternal and Child Health-IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (G.C.); (M.C.)
| | - Manola Comar
- Department of Advanced Microbiology Diagnosis and Translational Research, Institute for Maternal and Child Health-IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (G.C.); (M.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34100 Trieste, Italy;
| | - Chiara Ragazzon
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34100 Trieste, Italy;
| | - Riccardo Davanzo
- Neonatal Intensive Care Unit, Institute for Maternal and Child Health-IRCCS “Burlo Garofolo”, 34137 Trieste, Italy;
| | - Mariachiara Quadrifoglio
- Department of Obstetrics and Gynaecology, Institute for Maternal and Child Health-IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (F.S.); (M.Q.); (G.R.)
| | - Manuela Giangreco
- Epidemiology and Biostatistics Unit, Institute for Maternal and Child Health-IRCCS “Burlo Garofolo”, 34137 Trieste, Italy;
| | - Guglielmo Stabile
- Department of Obstetrics and Gynaecology, Institute for Maternal and Child Health-IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (F.S.); (M.Q.); (G.R.)
- Correspondence:
| | - Giuseppe Ricci
- Department of Obstetrics and Gynaecology, Institute for Maternal and Child Health-IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (F.S.); (M.Q.); (G.R.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34100 Trieste, Italy;
| |
Collapse
|
41
|
Bondt A, Dingess KA, Hoek M, van Rijswijck DMH, Heck AJR. A Direct MS-Based Approach to Profile Human Milk Secretory Immunoglobulin A (IgA1) Reveals Donor-Specific Clonal Repertoires With High Longitudinal Stability. Front Immunol 2021; 12:789748. [PMID: 34938298 PMCID: PMC8685336 DOI: 10.3389/fimmu.2021.789748] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/17/2021] [Indexed: 12/29/2022] Open
Abstract
Recently, a mass spectrometry-based approach was introduced to directly assess the IgG1 immunoglobulin clonal repertoires in plasma. Here we expanded upon this approach by describing a mass spectrometry-based technique to assess specifically the clonal repertoire of another important class of immunoglobulin molecules, IgA1, and show it is efficiently and robustly applicable to either milk or plasma samples. Focusing on two individual healthy donors, whose milk was sampled longitudinally during the first 16 weeks of lactation, we demonstrate that the total repertoire of milk sIgA1 is dominated by only 50-500 clones, even though the human body theoretically can generate several orders of magnitude more clones. We show that in each donor the sIgA1 repertoire only changes marginally and quite gradually over the monitored 16-week period of lactation. Furthermore, the observed overlap in clonal repertoires between the two individual donors is close to non-existent. Mothers provide protection to their newborn infants directly by the transfer of antibodies via breastfeeding. The approach introduced here, can be used to visualize the clonal repertoire transferred from mother to infant and to detect changes in-time in that repertoire adapting to changes in maternal physiology.
Collapse
Affiliation(s)
- Albert Bondt
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Center, Utrecht, Netherlands
| | - Kelly A Dingess
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Center, Utrecht, Netherlands
| | - Max Hoek
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Center, Utrecht, Netherlands
| | - Danique M H van Rijswijck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Center, Utrecht, Netherlands
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Center, Utrecht, Netherlands
| |
Collapse
|
42
|
Valcarce V, Stafford LS, Neu J, Cacho N, Parker L, Mueller M, Burchfield DJ, Li N, Larkin J. Detection of SARS-CoV-2-Specific IgA in the Human Milk of COVID-19 Vaccinated Lactating Health Care Workers. Breastfeed Med 2021; 16:1004-1009. [PMID: 34427487 DOI: 10.1089/bfm.2021.0122] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Background: In 2019, a deadly virus known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), responsible for coronavirus disease 2019 (COVID-19), emerged. In December 2020, two mRNA-based COVID-19 vaccines were approved for use in the United States, which provide immunity to those receiving the vaccine. Maternally derived antibodies are a key element of infants' immunity. Certain vaccines given to pregnant and lactating mothers provide immunity to infants through transmission across the placenta, umbilical cord (IgG), and human milk (IgA). Human milk produced by mothers with a history of COVID-19 infection contains SARS-CoV-2 IgA and IgG. The purpose of this study is to determine whether SARS-CoV-2-specific immunoglobulins are found in human milk after the COVID-19 vaccination, and to characterize the types of immunoglobulins present. Methods: This is a prospective observational study conducted at Shands Hospital, University of Florida, from December 2020 to March 2021. Twenty-two lactating health care workers who received the SARS-CoV-2 mRNA vaccine (Pfizer/BioNTech or Moderna) made up the sample group. Plasma and human milk were collected at three time points (prevaccination, post-first vaccine dose, and post-second vaccine dose). SARS-CoV-2-specific IgA and IgG in human milk and in plasma were measured by enzyme-linked immunosorbent assay (ELISA). Maternal demographics were compiled. Results: We found significant secretion of SARS-CoV-2-specific IgA and IgG in human milk and plasma after SARS-CoV-2 vaccination. Conclusions: Our results show that the mRNA-based COVID-19 vaccines induce SARS-CoV-2-specific IgA and IgG secretion in human milk. Further studies are needed to determine the duration of this immune response, its capacity to neutralize the COVID-19 virus, the transfer of passive immunity to breastfeeding infants, and the potential therapeutic use of human milk IgA to combat SARS-CoV-2 infections and COVID-19.
Collapse
Affiliation(s)
- Vivian Valcarce
- Department of Pediatrics and University of Florida, Gainesville, Florida, USA
| | - Lauren Stewart Stafford
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| | - Josef Neu
- Department of Pediatrics and University of Florida, Gainesville, Florida, USA
| | - Nicole Cacho
- Department of Pediatrics and University of Florida, Gainesville, Florida, USA
| | - Leslie Parker
- College of Nursing, University of Florida, Gainesville, Florida, USA
| | - Martina Mueller
- College of Nursing, Medical University of South Carolina, Charleston, South Carolina, USA
| | - David J Burchfield
- Department of Pediatrics and University of Florida, Gainesville, Florida, USA
| | - Nan Li
- College of Nursing, University of Florida, Gainesville, Florida, USA
| | - Joseph Larkin
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
43
|
Romero Ramírez DS, Lara Pérez MM, Carretero Pérez M, Suárez Hernández MI, Martín Pulido S, Pera Villacampa L, Fernández Vilar AM, Rivero Falero M, González Carretero P, Reyes Millán B, Roper S, García Bello MÁ. SARS-CoV-2 Antibodies in Breast Milk After Vaccination. Pediatrics 2021; 148:e2021052286. [PMID: 34408089 DOI: 10.1542/peds.2021-052286] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/12/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Passive and active immunity transfer through human milk (HM) constitutes a key element in the infant's developing immunity. Certain infectious diseases and vaccines have been described to induce changes in the immune components of HM. METHODS We conducted a prospective cohort single-institution study from February 2 to April 4, 2021. Women who reported to be breastfeeding at the time of their coronavirus disease 2019 (COVID-19) vaccination were invited to participate. Blood and milk samples were collected on day 14 after their second dose of the vaccine. Immunoglobulin G (IgG) antibodies against nucleocapsid protein as well as IgG, immunoglobulin M and immunoglobulin A (IgA) antibodies against the spike 1 protein receptor-binding domain against severe acute respiratory syndrome coronavirus 2 (anti-SARS-CoV-2 RBD-S1) were analyzed in both serum and HM samples. RESULTS Most of the participants (ie, 94%) received the BNT162b2 messenger RNA COVID-19 vaccine. The mean serum concentration of anti-SARS-CoV-2 RBD-S-IgG antibodies in vaccinated individuals was 3379.6 ± 1639.5 binding antibody units per mL. All vaccinated study participants had anti-SARS-CoV-2 RBD-S1-IgG, and 89% of them had anti-SARS-CoV-2 RBD-S-IgA in their milk. The antibody concentrations in the milk of mothers who were breastfeeding 24 months were significantly higher than in mothers with breastfeeding periods <24 months (P < .001). CONCLUSIONS We found a clear association between COVID-19 vaccination and specific immunoglobulin concentrations in HM. This effect was more pronounced when lactation periods exceeded 23 months. The influence of the lactation period on immunoglobulins was specific and independent of other variables.
Collapse
Affiliation(s)
- Dolores Sabina Romero Ramírez
- Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- Contributed equally as co-first authors
| | - María Magdalena Lara Pérez
- Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- Contributed equally as co-first authors
| | | | | | - Saúl Martín Pulido
- Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | | | | | - Mónica Rivero Falero
- Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | | | - Beatriz Reyes Millán
- Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Sabine Roper
- Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | | |
Collapse
|
44
|
Dawod B, Marshall JS, Azad MB. Breastfeeding and the developmental origins of mucosal immunity: how human milk shapes the innate and adaptive mucosal immune systems. Curr Opin Gastroenterol 2021; 37:547-556. [PMID: 34634003 PMCID: PMC11451935 DOI: 10.1097/mog.0000000000000778] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Breastfeeding provides passive immunity while the neonatal immune system matures, and may also protect against chronic immune-mediated conditions long after weaning. This review summarizes current knowledge and new discoveries about human milk and mucosal immunity. RECENT FINDINGS New data suggest that certain microbes in maternal milk may seed and shape the infant gut microbiota, which play a key role in regulating gut barrier integrity and training the developing immune system. Human milk oligosaccharides, best known for their prebiotic functions, have now been shown to directly modulate gene expression in mast and goblet cells in the gastrointestinal tract. Epidemiologic data show a reduced risk of peanut sensitization among infants breastfed by peanut-consuming mothers, suggesting a role for milk-borne food antigens in tolerance development. Cross-fostering experiments in mice suggest the soluble Toll-like receptor 2, found in human milk, may be critical in this process. Finally, interest in human milk antibodies surged during the pandemic with the identification of neutralizing severe acute respiratory syndrome coronavirus 2 antibodies in maternal milk following both natural infection and vaccination. SUMMARY Human milk provides critical immune protection and stimulation to breastfed infants. Understanding the underlying mechanisms could identify new therapeutic targets and strategies for disease prevention across the lifespan.
Collapse
Affiliation(s)
- Bassel Dawod
- Department of Pathology
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia
| | - Jean S. Marshall
- Department of Pathology
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia
| | - Meghan B. Azad
- Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba
- Department of Pediatrics and Child Health
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
45
|
Charepe N, Gonçalves J, Juliano AM, Lopes DG, Canhão H, Soares H, Serrano EF. COVID-19 mRNA vaccine and antibody response in lactating women: a prospective cohort study. BMC Pregnancy Childbirth 2021; 21:632. [PMID: 34535094 PMCID: PMC8447894 DOI: 10.1186/s12884-021-04051-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 08/12/2021] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Immunological protection via breastfeeding is well known. The immunological profile of human milk changes during lactation. No clinical trials have been conducted in lactating women with the newest mRNA vaccines against SARS- CoV-2. A Few studies have shown the presence of antibodies in breastmilk after vaccination. The aim of this work is to study possible antibodies transfer via breastmilk and also the immunological characteristics of lactating women compared to non-lactating women, after using the BNT162b2 Pfizer vaccine. METHODS This is a prospective cohort study with a convenience homogenous sample of 24 healthcare workers (14 lactating and 10 non-lactating women) enrolled at the time of COVID-19 vaccination. Clinical data was registered in a questionnaire. Titers of SARS-CoV-2 spike IgG, IgA and IgM were quantified in post vaccination blood and human milk. Antibody quantification was performed by an in-house ELISA to SARS-CoV-2 trimeric spike protein. RESULTS All women showed immunity after vaccination with positive antibodies for IgM, IgA and IgG antibodies. The dominant serum antibody response was IgG. Modest levels of antibodies in breastmilk of lactating mothers were observed in this study, especially IgG in 42.9%. There was a moderate association between higher titers of IgG and a longer duration of breastfeeding (R= 0.55, p=0.041). CONCLUSIONS Evidence of antibody transfer in human milk after COVID-19 vaccination is scarce. The presence of antibodies in human milk is reported, but immunization through breastfeeding is still to be established.
Collapse
Affiliation(s)
- Nadia Charepe
- Centro Hospitalar Universitário de Lisboa Central (CHULC), Lisboa, Portugal.
- Comprehensive Health Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal.
| | - Juliana Gonçalves
- Human Immunobiology and Pathogenesis Laboratory, Lisbon, Portugal
- CEDOC-Chronic Diseases Research Center NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisbon, Portugal
| | - A Margarida Juliano
- Human Immunobiology and Pathogenesis Laboratory, Lisbon, Portugal
- CEDOC-Chronic Diseases Research Center NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisbon, Portugal
| | - David G Lopes
- Comprehensive Health Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
- EpiDoC Unit, CEDOC, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Helena Canhão
- Centro Hospitalar Universitário de Lisboa Central (CHULC), Lisboa, Portugal
- Comprehensive Health Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
- EpiDoC Unit, CEDOC, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Helena Soares
- Comprehensive Health Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
- Human Immunobiology and Pathogenesis Laboratory, Lisbon, Portugal
- CEDOC-Chronic Diseases Research Center NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisbon, Portugal
| | - E Fátima Serrano
- Centro Hospitalar Universitário de Lisboa Central (CHULC), Lisboa, Portugal
- Comprehensive Health Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| |
Collapse
|
46
|
Guida M, Terracciano D, Cennamo M, Aiello F, La Civita E, Esposito G, Gargiulo V, Maruotti GM, Portella G, Sarno L. COVID-19 Vaccine mRNABNT162b2 Elicits Human Antibody Response in Milk of Breastfeeding Women. Vaccines (Basel) 2021; 9:vaccines9070785. [PMID: 34358201 PMCID: PMC8310008 DOI: 10.3390/vaccines9070785] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/06/2021] [Accepted: 07/12/2021] [Indexed: 12/23/2022] Open
Abstract
Objective: The objective of this research is to demonstrate the release of SARS-CoV-2 Spike (S) antibodies in human milk samples obtained by patients who have been vaccinated with mRNABNT162b2 vaccine. Methods: Milk and serum samples were collected in 10 volunteers 20 days after the first dose and 7 seven days after the second dose of the mRNABNT162b2 vaccine. Anti-SARS-CoV-2 S antibodies were measured by the Elecsys® Anti-SARS-CoV-2 S ECLIA assay (Roche Diagnostics AG, Rotkreuz, Switzerland), a quantitative electrochemiluminescence immunometric method. Results: At first sample, anti-SARS-CoV-2 S antibodies were detected in all serum samples (103.9 ± 54.9 U/mL) and only in two (40%) milk samples with a low concentration (1.2 ± 0.3 U/mL). At the second sample, collected 7 days after the second dose, anti-SARS-CoV-2 S antibodies were detected in all serum samples (3875.7 ± 3504.6 UI/mL) and in all milk samples (41.5 ± 47.5 UI/mL). No correlation was found between the level of serum and milk antibodies; the milk antibodies/serum antibodies ratio was on average 2% (range: 0.2–8.4%). Conclusion: We demonstrated a release of anti-SARS-CoV-2 S antibodies in the breast milk of women vaccinated with mRNABNT162b2. Vaccinating breastfeeding women could be a strategy to protect their infants from COVID-19 infection.
Collapse
Affiliation(s)
- Maurizio Guida
- Department of Neurosciences, Reproductive Science and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (M.G.); (F.A.); (G.M.M.); (L.S.)
| | - Daniela Terracciano
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (M.C.); (E.L.C.); (G.P.)
- Correspondence: ; Tel.: +39-0817463617
| | - Michele Cennamo
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (M.C.); (E.L.C.); (G.P.)
| | - Federica Aiello
- Department of Neurosciences, Reproductive Science and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (M.G.); (F.A.); (G.M.M.); (L.S.)
| | - Evelina La Civita
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (M.C.); (E.L.C.); (G.P.)
| | - Gennaro Esposito
- Department of Public Health, University of Naples “Federico II”, 80131 Naples, Italy;
| | - Valentina Gargiulo
- Department of Mother and Child, University Hospital Federico II, 80131 Naples, Italy;
| | - Giuseppe M. Maruotti
- Department of Neurosciences, Reproductive Science and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (M.G.); (F.A.); (G.M.M.); (L.S.)
| | - Giuseppe Portella
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (M.C.); (E.L.C.); (G.P.)
| | - Laura Sarno
- Department of Neurosciences, Reproductive Science and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (M.G.); (F.A.); (G.M.M.); (L.S.)
| |
Collapse
|
47
|
Noel G, In JG, Lemme-Dumit JM, DeVine LR, Cole RN, Guerrerio AL, Campbell JD, Kovbasnjuk O, Pasetti MF. Human Breast Milk Enhances Intestinal Mucosal Barrier Function and Innate Immunity in a Healthy Pediatric Human Enteroid Model. Front Cell Dev Biol 2021; 9:685171. [PMID: 34327199 PMCID: PMC8313895 DOI: 10.3389/fcell.2021.685171] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/18/2021] [Indexed: 12/20/2022] Open
Abstract
Breastfeeding has been associated with long lasting health benefits. Nutrients and bioactive components of human breast milk promote cell growth, immune development, and shield the infant gut from insults and microbial threats. The molecular and cellular events involved in these processes are ill defined. We have established human pediatric enteroids and interrogated maternal milk's impact on epithelial cell maturation and function in comparison with commercial infant formula. Colostrum applied apically to pediatric enteroid monolayers reduced ion permeability, stimulated epithelial cell differentiation, and enhanced tight junction function by upregulating occludin. Breast milk heightened the production of antimicrobial peptide α-defensin 5 by goblet and Paneth cells, and modulated cytokine production, which abolished apical release of pro-inflammatory GM-CSF. These attributes were not found in commercial infant formula. Epithelial cells exposed to breast milk elevated apical and intracellular pIgR and enabled maternal IgA translocation. Proteomic data revealed a breast milk-induced molecular pattern associated with tissue remodeling and homeostasis. Using a novel ex vivo pediatric enteroid model, we have identified distinct cellular and molecular events involved in human milk-mediated improvement of human intestinal physiology and immunity.
Collapse
Affiliation(s)
- Gaelle Noel
- Department of Pediatrics, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Julie G. In
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of New Mexico Health Science Center, Albuquerque, NM, United States
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jose M. Lemme-Dumit
- Department of Pediatrics, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Lauren R. DeVine
- Department of Biological Chemistry, Johns Hopkins Mass Spectrometry and Proteomics Facility, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Robert N. Cole
- Department of Biological Chemistry, Johns Hopkins Mass Spectrometry and Proteomics Facility, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Anthony L. Guerrerio
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - James D. Campbell
- Department of Pediatrics, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Olga Kovbasnjuk
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of New Mexico Health Science Center, Albuquerque, NM, United States
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Marcela F. Pasetti
- Department of Pediatrics, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
48
|
Immune Response to Vaccination against COVID-19 in Breastfeeding Health Workers. Vaccines (Basel) 2021; 9:vaccines9060663. [PMID: 34204501 PMCID: PMC8235492 DOI: 10.3390/vaccines9060663] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/10/2021] [Accepted: 06/15/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Initially, there were no data on the safety of COVID-19 vaccines in lactating women. The aim of our study was to evaluate the immune response to COVID-19 vaccinations in breastfeeding women. Methods: The study included 32 breastfeeding women who, regardless of the study, had decided to be vaccinated. Maternal serum and breast milk samples were simultaneously collected on days 8 ± 1, 22 ± 2, 29 ± 3, and 43 ± 4 after the first dose of the vaccine. The immune response was assessed by determining the presence of anti-SARS-CoV-2 IgG and IgA. Results: The breast milk IgG level was detectable (6.50 ± 6.74, median 4.7, and maximum 34.2 BAU/mL) and highly correlated to serum IgG level (rS 0.89; p < 0.001). The breast milk ratio of IgA to the cut-off value was higher in serum IgA-positive (4.18 ± 3.26, median 2.8, and maximum >10) than in serum IgA-negative women (0.56 ± 0.37, median 0.5, and maximum 1.6; p < 0.001). The highest concentrations of serum and breast milk antibodies were observed on day 29 ± 3 with a decrease on day 43 ± 4. Conclusion: The immune response to the vaccination against SARS-CoV-2 is strongest 7 ± 3 days after the second dose of the vaccine. Lactating mothers breastfeeding their children after vaccination against SARS-CoV-2 may transfer antibodies to their infant.
Collapse
|
49
|
Lackey KA, Fehrenkamp BD, Pace RM, Williams JE, Meehan CL, McGuire MA, McGuire MK. Breastfeeding Beyond 12 Months: Is There Evidence for Health Impacts? Annu Rev Nutr 2021; 41:283-308. [PMID: 34115518 DOI: 10.1146/annurev-nutr-043020-011242] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Because breastfeeding provides optimal nutrition and other benefits for infants (e.g., lower risk of infectious disease) and benefits for mothers (e.g., less postpartum bleeding), health organizations recommend that healthy infants be exclusively breastfed for 4 to 6 months in the United States and 6 months internationally. Recommendations related to how long breastfeeding should continue, however, are inconsistent. The objective of this article is to review the literature related to evidence for benefits of breastfeeding beyond 1 year for mothers and infants. In summary, human milk represents a good source of nutrients and immune components beyond 1 year. Some studies point toward lower infant mortality in undernourished children breastfed for >1 year, and prolonged breastfeeding increases interbirth intervals. Data on other outcomes (e.g., growth, diarrhea, obesity, and maternal weight loss) are inconsistent, often lacking sufficient control for confounding variables. There is a substantial need for rigorous, prospective, mixed-methods, cross-cultural research on this topic. Expected final online publication date for the Annual Review of Nutrition, Volume 41 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Kimberly A Lackey
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, Idaho 83844, USA;
| | - Bethaney D Fehrenkamp
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, Idaho 83844, USA;
| | - Ryan M Pace
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, Idaho 83844, USA;
| | - Janet E Williams
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, Idaho 83844, USA
| | - Courtney L Meehan
- Department of Anthropology, Washington State University, Pullman, Washington 99164, USA
| | - Mark A McGuire
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, Idaho 83844, USA
| | - Michelle K McGuire
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, Idaho 83844, USA;
| |
Collapse
|
50
|
Hase K, Stahmer L, Shammas H, Peter C, Bohnhorst B, Das AM. Analysis of Sirtuin 1 and Sirtuin 3 at Enzyme and Protein Levels in Human Breast Milk during the Neonatal Period. Metabolites 2021; 11:metabo11060348. [PMID: 34072556 PMCID: PMC8229955 DOI: 10.3390/metabo11060348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 12/13/2022] Open
Abstract
Breast feeding is regarded as the preferred nutrition modality for children during the first few months of life. It not only furthers growth and development but also is supposed to impact later life. The first 1000 days are regarded as a critical window for development, even beyond infancy. The physiological basis underlying this beneficial effect is not clear. Sirtuins are important regulatory proteins of metabolism and are supposed to play a critical role in ageing and longevity as well as in diseases. In the present study, we developed novel methods to assay sirtuin 1 and sirtuin 3 at enzyme activity (via fluorometry) and protein levels (by Western blot) in the aqueous phase and in the cell pellet of human breast milk and assessed the impact of ongoing lactation during the neonatal period. Sirtuin activities in the aqueous phase were negatively correlated with the duration of lactation in the neonatal period. There was no correlation of sirtuin activities in the cell pellet with the duration of lactation. The amounts of sirtuin 1 and sirtuin 3 measured by Western blot were negatively correlated with the lactation period.
Collapse
Affiliation(s)
- Kristina Hase
- Clinic for Pediatric Kidney-, Liver- and Metabolic Diseases, Hannover Medical School, 30625 Hannover, Germany; (K.H.); (L.S.); (H.S.)
| | - Laura Stahmer
- Clinic for Pediatric Kidney-, Liver- and Metabolic Diseases, Hannover Medical School, 30625 Hannover, Germany; (K.H.); (L.S.); (H.S.)
| | - Hadeel Shammas
- Clinic for Pediatric Kidney-, Liver- and Metabolic Diseases, Hannover Medical School, 30625 Hannover, Germany; (K.H.); (L.S.); (H.S.)
| | - Corinna Peter
- Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany; (C.P.); (B.B.)
| | - Bettina Bohnhorst
- Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany; (C.P.); (B.B.)
| | - Anibh Martin Das
- Clinic for Pediatric Kidney-, Liver- and Metabolic Diseases, Hannover Medical School, 30625 Hannover, Germany; (K.H.); (L.S.); (H.S.)
- Correspondence: ; Tel.: +49-511-532-3273 (ext. 3220)
| |
Collapse
|