1
|
Farooq U, Ahmed S, Liu G, Jiang X, Yang H, Ding J, Ali M. Biochemical properties of sheep colostrum and its potential benefits for lamb survival: a review. Anim Biotechnol 2024; 35:2320726. [PMID: 38436999 DOI: 10.1080/10495398.2024.2320726] [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: 03/05/2024]
Abstract
Colostrum is the initial secretion of the mammary glands following parturition, which offers main food, protection, and biological active substances for the new born. The most threatening episode of neonate's life is the initial two weeks after birth. This period is associated with high neonatal mortality and morbidity. These worthwhile losses lead to a poor prolificacy rate, low profitability, and ultimately poor performance in animal production. Hence, both diseases and mortality cause valuable losses in terms of production and economic losses. The survival of neonate is correlated with their immune status and passive immune transfer (PIT). Colostrum provides the primary source of nutrition and immunity (PIT) that protects neonates against infections. It must be given as soon as possible after birth since its immunoglobulins are absorbed within the first 16-27 hours after birth, ideally within 2-4 hours. As a result, immunoglobulin (PIT) is the most important component of distressing infectious immunity, and a passable concentration of immunoglobulin in the blood of newborn lambs is linked to their health and survival rate. In this review, we summarized the importance of colostrum in early life and its association with neonatal lamb's survival, profitability and productivity of sheep farming.
Collapse
Affiliation(s)
- Umar Farooq
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Livestock and Dairy Development Department Lahore, Punjab, Pakistan
| | - Sohail Ahmed
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Laboratory of Small Ruminant Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Guiqiong Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Laboratory of Small Ruminant Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xunping Jiang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Laboratory of Small Ruminant Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Huiguo Yang
- Xinjiang Academy of Animal Sciences, Urumuqi, China
| | - Jianping Ding
- Anhui Anxin (Woyang) Animal Husbandry Development Co., Ltd., Bozhou, China
| | - Mehboob Ali
- State key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
2
|
Furuta S. Microbiome-Stealth Regulator of Breast Homeostasis and Cancer Metastasis. Cancers (Basel) 2024; 16:3040. [PMID: 39272898 PMCID: PMC11394247 DOI: 10.3390/cancers16173040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Cumulative evidence attests to the essential roles of commensal microbes in the physiology of hosts. Although the microbiome has been a major research subject since the time of Luis Pasteur and William Russell over 140 years ago, recent findings that certain intracellular bacteria contribute to the pathophysiology of healthy vs. diseased tissues have brought the field of the microbiome to a new era of investigation. Particularly, in the field of breast cancer research, breast-tumor-resident bacteria are now deemed to be essential players in tumor initiation and progression. This is a resurrection of Russel's bacterial cause of cancer theory, which was in fact abandoned over 100 years ago. This review will introduce some of the recent findings that exemplify the roles of breast-tumor-resident microbes in breast carcinogenesis and metastasis and provide mechanistic explanations for these phenomena. Such information would be able to justify the utility of breast-tumor-resident microbes as biomarkers for disease progression and therapeutic targets.
Collapse
Affiliation(s)
- Saori Furuta
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, 2500 MetroHealth Drive, Cleveland, OH 44109, USA;
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
3
|
Kroll KW, Hueber B, Balachandran H, Afifi A, Manickam C, Nettere D, Pollara J, Hudson A, Woolley G, Ndhlovu LC, Reeves RK. FcαRI (CD89) is upregulated on subsets of mucosal and circulating NK cells and regulates IgA-class specific signaling and functions. Mucosal Immunol 2024; 17:692-699. [PMID: 38677592 PMCID: PMC11323182 DOI: 10.1016/j.mucimm.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 03/27/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024]
Abstract
Immunoglobulin A (IgA) is the predominant mucosal antibody class with both anti- and pro-inflammatory roles1-3. However, the specific role of the IgA receptor cluster of differentiation (CD)89, expressed by a subset of natural killer (NK) cells, is poorly explored. We found that CD89 protein expression on circulating NK cells is infrequent in humans and rhesus macaques, but transcriptomic analysis showed ubiquitous CD89 expression, suggesting an inducible phenotype. Interestingly, CD89+ NK cells were more frequent in cord blood and mucosae, indicating a putative IgA-mediated NK cell function in the mucosae and infant immune system. CD89+ NK cells signaled through upregulated CD3 zeta chain (CD3ζ), spleen tyrosine kinase (Syk), zeta chain-associated protein kinase 70 (ZAP70), and signaling lymphocytic activation molecule family 1 (SLAMF1), but also showed high expression of inhibitory receptors such as killer cell lectin-like receptor subfamily G (KLRG1) and reduced activating NKp46 and NKp30. CD89-based activation or antibody-mediated cellular cytotoxicity with monomeric IgA1 reduced NK cell functions, while antibody-mediated cellular cytotoxicity with combinations of IgG and IgA2 was enhanced compared to IgG alone. These data suggest that functional CD89+ NK cells survey mucosal sites, but CD89 likely serves as regulatory receptor which can be further modulated depending on IgA and IgG subclass. Although the full functional niche of CD89+ NK cells remains unexplored, these intriguing data suggest the CD89 axis could represent a novel immunotherapeutic target in the mucosae or early life.
Collapse
Affiliation(s)
- Kyle W Kroll
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, USA; Duke University School of Medicine, Durham, North Carolina, USA; Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Brady Hueber
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, USA; Duke University School of Medicine, Durham, North Carolina, USA; Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Harikrishnan Balachandran
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, USA; Duke University School of Medicine, Durham, North Carolina, USA; Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Ameera Afifi
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, USA; Duke University School of Medicine, Durham, North Carolina, USA; Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Cordelia Manickam
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, USA; Duke University School of Medicine, Durham, North Carolina, USA; Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Danielle Nettere
- Duke University School of Medicine, Durham, North Carolina, USA; Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Justin Pollara
- Duke University School of Medicine, Durham, North Carolina, USA; Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Andrew Hudson
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, USA; Duke University School of Medicine, Durham, North Carolina, USA; Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Griffin Woolley
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, USA; Duke University School of Medicine, Durham, North Carolina, USA; Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Lishomwa C Ndhlovu
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York City, New York, USA
| | - R Keith Reeves
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, USA; Duke University School of Medicine, Durham, North Carolina, USA; Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA.
| |
Collapse
|
4
|
Jaquish A, Phung E, Gong X, Baldominos-Flores P, Galvan-Pena S, Bursulaya I, Magill I, Bertrand K, Chambers C, Agudo J, Mathis D, Benoist C, Ramanan D. Expansion of mammary intraepithelial lymphocytes and intestinal inputs shape T cell dynamics in lactogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.09.602739. [PMID: 39026711 PMCID: PMC11257640 DOI: 10.1101/2024.07.09.602739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Pregnancy brings about profound changes to the mammary gland in preparation for lactation. Changes in immunocyte populations that accompany this rapid remodeling are incompletely understood. We comprehensively analyzed mammary T cells through all parous stages, revealing a marked increase in CD4+ and CD8+ T effector cells in late pregnancy and lactation. T cell expansion was partly dependent on microbial signals and included an increase in TCRαβ+CD8αα+ cells with strong cytotoxic markers, located in the epithelium, that resemble intraepithelial lymphocytes of mucosal tissues. This relationship was substantiated by demonstrating T cell migration from gut to mammary gland in late pregnancy, by TCR clonotypes shared by intestine and mammary tissue in the same mouse, including intriguing gut TCR families. Putative counterparts of CD8αα+ IELs were found in human milk. Mammary T cells are thus poised to manage the transition from a non-mucosal tissue to a mucosal barrier during lactogenesis.
Collapse
|
5
|
Liu J, Yang DA, Qu H, Liu D, Huang K. Bacillus subtilis Feed Supplementation Combined with Oral E. coli Immunization in Sows as a Tool to Reduce Neonatal Diarrhea in Piglets. Animals (Basel) 2024; 14:1978. [PMID: 38998090 PMCID: PMC11240787 DOI: 10.3390/ani14131978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 06/30/2024] [Accepted: 07/02/2024] [Indexed: 07/14/2024] Open
Abstract
To investigate the effects of B. subtilis on the specific immune response of lactating sows to E. coli and the diarrhea rate in suckling piglets, thirty large white sows with similar farrowing dates were randomly divided into two groups: a feedback feeding (i.e., feeding a homogenate of intestinal contents and tissues from E. coli-infected piglets to sows; FB) group and a feedback feeding with B. subtilis (FB + BS) group. Serum, colostrum, and intestinal tissues from sows and piglets were collected to assess the immune response and intestinal barrier function at weaning. T and B cells from Peyer's patches (PPs) and mesenteric lymph nodes (MLNs) in lactating mice (with treatments consistent with the sows') were isolated to explore the underlying mechanism. The results showed that, compared with the FB group, the reproductive performance of sows and the growth performance of their offspring were effectively improved in the FB + BS group. Moreover, the levels of IgG/IgA and those of IgG/IgA against E. coli in the serum and colostrum of sows in the FB+BS group were increased (p < 0.05). Meanwhile, the ratio of CD4+/CD8+, CD4+CXCR5+PD1+, and B220+IgA+ cells in MLNs and PPs, and the IgA levels in the mammary glands of mice, were also increased in the FB + BS group (p < 0.05). Notably, in suckling piglets in the FB + BS group, the diarrhea rate was decreased (p < 0.05), and the intestinal barrier function and intestinal flora composition at weaning were significantly improved. Overall, these results indicated that B. subtilis feed supplementation combined with feedback feeding in pregnant and lactating sows can reduce diarrhea in suckling piglets by enhancing the maternal immune response against E. coli and intestinal barrier function in their offspring, improving survival rates and pre-weaning growth.
Collapse
Affiliation(s)
- Jianxin Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing 210095, China
| | - Danchen Aaron Yang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Haobo Qu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing 210095, China
| | - Dandan Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing 210095, China
| | - Kehe Huang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
6
|
Hu Y, Wu X, Zhou L, Liu J. Which is the optimal choice for neonates' formula or breast milk? NATURAL PRODUCTS AND BIOPROSPECTING 2024; 14:21. [PMID: 38488905 PMCID: PMC10942964 DOI: 10.1007/s13659-024-00444-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 02/28/2024] [Indexed: 03/17/2024]
Abstract
The incidence of prematurity has been increasing since the twenty-first century. Premature neonates are extremely vulnerable and require a rich supply of nutrients, including carbohydrates, proteins, docosahexaenoic acid (DHA), arachidonic acid (ARA) and others. Typical breast milk serves as the primary source for infants under six months old to provide these nutrients. However, depending on the individual needs of preterm infants, a more diverse and intricate range of nutrients may be necessary. This paper provides a comprehensive review of the current research progress on the physical and chemical properties, biological activity, function, and structure of breast milk, as well as explores the relationship between the main components of milk globular membrane and infant growth. Additionally, compare the nutritional composition of milk from different mammals and newborn milk powder, providing a comprehensive understanding of the differences in milk composition and detailed reference for meeting daily nutritional needs during lactation.
Collapse
Affiliation(s)
- Yueqi Hu
- National Demonstration Center for Experimental Ethnopharmacology Education, School of Pharmaceutical Sciences, South-Central MinZu University, Wuhan, 430074, People's Republic of China
| | - Xing Wu
- National Demonstration Center for Experimental Ethnopharmacology Education, School of Pharmaceutical Sciences, South-Central MinZu University, Wuhan, 430074, People's Republic of China
| | - Li Zhou
- National Demonstration Center for Experimental Ethnopharmacology Education, School of Pharmaceutical Sciences, South-Central MinZu University, Wuhan, 430074, People's Republic of China.
| | - Jikai Liu
- National Demonstration Center for Experimental Ethnopharmacology Education, School of Pharmaceutical Sciences, South-Central MinZu University, Wuhan, 430074, People's Republic of China.
| |
Collapse
|
7
|
Urrete J, Mitra T, Boland BS, Bertrand K, Chambers C, Rivera-Nieves J. Vedolizumab Does Not Affect Antibody Secreting Cell Recruitment to the Lactating Mammary Gland of Mothers With Inflammatory Bowel Disease. Inflamm Bowel Dis 2024:izae023. [PMID: 38334263 DOI: 10.1093/ibd/izae023] [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: 12/07/2023] [Indexed: 02/10/2024]
Abstract
Lay Summary
Despite a known role for α4β7 and MAdCAM-1 for the recruitment of antibody secreting cells to the lactating mammary gland, vedolizumab which targets integrin α4β7 did not lower breastmilk IgA in lactating mothers with IBD receiving the drug. It is likely that antibody secreting cells alternatively employ α4β1 to arrest on VCAM-1 also expressed by the lactating mammary gland.
Collapse
Affiliation(s)
- Josef Urrete
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
| | - Taniya Mitra
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
- GI Section, San Diego VA Medical Center (SDVAMC), San Diego, CA, USA
| | - Brigid S Boland
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
| | - Kerri Bertrand
- Department of Pediatrics, Division of Environmental Science and Health, University of California San Diego, La Jolla, CA, USAñ
| | - Christina Chambers
- Department of Pediatrics, Division of Environmental Science and Health, University of California San Diego, La Jolla, CA, USAñ
| | - Jesús Rivera-Nieves
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
- GI Section, San Diego VA Medical Center (SDVAMC), San Diego, CA, USA
| |
Collapse
|
8
|
Ahmed S, Jiang X, Liu G, Yang H, Sadiq A, Yi D, Farooq U, Yiyu S, Zubair M. The protective role of maternal genetic immunization on maternal-fetal health and welfare. Int J Gynaecol Obstet 2023; 163:763-777. [PMID: 37218379 DOI: 10.1002/ijgo.14853] [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: 03/13/2023] [Accepted: 04/28/2023] [Indexed: 05/24/2023]
Abstract
Pregnancy is a critical period associated with alterations in physiologic, biologic, and immunologic processes, which can affect maternal-fetal health through development of several infectious diseases. At birth, neonates have an immature immune system that makes them more susceptible to severe viral infections and diseases. For this reason, different maternal nutritional and immunization interventions have been used to improve the immune and health status of the mother and her neonate through passive immunity. Here, we reviewed the protective role of maternal immunization with different types of vaccines, especially genetic vaccines, during pregnancy in maternal-fetal health, immune response, colostrum quality, immune response, and anti-oxidative status. For this purpose, we have used different scientific databases (PubMed and Google Scholar) and other official web pages. We customized the search period range from the year 2000 to 2023 using the key words "maternal immunization" OR "gestation period/pregnancy" OR "genetic vaccination" OR "maternal-fetal health" OR "micronutrients" OR "neonatal immunity" "oxidative stress" OR "colostrum quality". The evidence demonstrated that inactivated or killed vaccines produced significant immune protection in the mother and fetus. Furthermore, most recent studies have suggested that the use of genetic vaccines (mRNA and DNA) during pregnancy is efficient at triggering the immune response in mother and neonate without the risk of undesired pregnancy outcomes. However, factors such as maternal redox balance, nutritional status, and the timing of immunization play essential roles in regulating immune response inflammatory status, antioxidant capacity, and the welfare of both the pregnant mother and her newborn.
Collapse
Affiliation(s)
- Sohail Ahmed
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Laboratory of Sheep and Goat Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xunping Jiang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Laboratory of Sheep and Goat Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Smart Farming for Agricultural Animals, Wuhan, China
| | - Guiqiong Liu
- Laboratory of Sheep and Goat Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Huiguo Yang
- Xinjiang Academy of Animal Sciences, Urumqi, China
| | - Amber Sadiq
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, Pakistan
| | - Ding Yi
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Laboratory of Sheep and Goat Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Umar Farooq
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
| | - Sha Yiyu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Laboratory of Sheep and Goat Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Muhammad Zubair
- Department of Veterinary Clinical Sciences, University of Poonch, Rawalakot, Pakistan
| |
Collapse
|
9
|
Debbag R, Torres JR, Falleiros-Arlant LH, Avila-Aguero ML, Brea-del Castillo J, Gentile A, Saez-Llorens X, Mascarenas A, Munoz FM, Torres JP, Vazquez L, Safadi MA, Espinal C, Ulloa-Gutierrez R, Pujadas M, Lopez P, López-Medina E, Ramilo O. Are the first 1,000 days of life a neglected vital period to prevent the impact on maternal and infant morbimortality of infectious diseases in Latin America? Proceedings of a workshop of experts from the Latin American Pediatric Infectious Diseases Society, SLIPE. Front Pediatr 2023; 11:1297177. [PMID: 38098643 PMCID: PMC10720332 DOI: 10.3389/fped.2023.1297177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/15/2023] [Indexed: 12/17/2023] Open
Abstract
While the first 1,000 days of life are a critical period in child's development, limited information on the main determinants affecting this period in the Latin America and the Caribbean (LAC) region is available. Therefore, the Latin American Pediatric Infectious Diseases Society (SLIPE) held an ad hoc workshop in May 2022 with an expert panel designed to analyze the main factors impacting the development of childhood in the region during this period and the main causes of maternal infant morbimortality. The aim was to identify priorities, generate recommendations, and advise practical actions to improve this situation. Considerations were made about the challenges involved in bridging the gap that separates the region from more developed countries regarding an optimal early childhood and maternal care. Extensive discussion was conducted to reach consensus recommendations on general strategies intended to reduce maternal and infant mortality associated with infections and immune-preventable diseases during the first 1,000 days of life in LAC.
Collapse
Affiliation(s)
- Roberto Debbag
- President of Sociedad Latinoamericana de Infectología Pediátrica, SLIPE, Buenos Aires, Argentina
| | - Jaime R. Torres
- Infectious Diseases Section, Tropical Medicine Institute, Universidad Central De Venezuela, Caracas, Venezuela
| | - Luiza H. Falleiros-Arlant
- Department of Children’s Health, Faculdade De Medicina, Universidade Metropolitana De Santos, Santos, Brazil
| | - Maria L. Avila-Aguero
- Infectious Diseases Service, Hospital Nacional De Niños “Dr. Carlos Sáenz Herrera”, Caja Costarricense De Seguro Social (CCSS), San José, Costa Rica
- Affiliated Researcher Center for Infectious Disease Modeling and Analysis (CIDMA) at Yale University, New Haven, CT, United States
| | - Jose Brea-del Castillo
- Associated Researcher, Investigador Asociado Hospital Dr. Hugo Mendoza, Santo Domingo, Republic Dominicana
| | - Angela Gentile
- Epidemiology Department, Hospital de Niños “Ricardo Gutiérrez”, Buenos Aires University, Buenos Aires, Argentina
| | - Xavier Saez-Llorens
- Head of Infectious Diseases and Director of Clinical Research, Hospital del Niño “Dr. José Renán Esquivel”, Panama City, Panama
| | - Abiel Mascarenas
- Department of Pediatric Infectious Diseases, Hospital Universitario “José E. Gonzalez”, Universidad Autónoma De Nuevo León, Nuevo Leon, México
| | - Flor M. Munoz
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Juan P. Torres
- Department of Pediatrics and Children Surgery, Universidad de Chile, Santiago, Chile
| | - Liliana Vazquez
- Pediatric Infectious Diseases, Clinica y Maternidad Suizo Argentina, Sanatorio Finochietto, Buenos Aires, Argentina
| | - Marco A. Safadi
- Department of Pediatrics, Faculda de de Ciências Médicas da Santa Casa de São Paulo, Sao Paulo, Brazil
| | - Carlos Espinal
- Global Health Consortium, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States
| | - Rolando Ulloa-Gutierrez
- Infectious Diseases Service, Hospital Nacional De Niños “Dr. Carlos Sáenz Herrera”, Caja Costarricense De Seguro Social (CCSS), San José, Costa Rica
| | - Monica Pujadas
- Department of Epidemiology and Pediatric Infectious Diseases, Centro Hospitalario Pereira Rossell, Faculty of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Pio Lopez
- Department of Pediatrics, Hospital Universitario del Valle, Cali, Colombia
| | - Eduardo López-Medina
- Centro de Estudios en Infectología Pediátrica CEIP, Department of Pediatrics, Universidad del Valle, Clinica Imbanaco Grupo Quironsalud, Cali, Colombia
| | - Octavio Ramilo
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN, United States
| |
Collapse
|
10
|
Liu W, Jiang H, Liu X, Zheng Y, Liu Y, Pan F, Yu F, Li Z, Gu M, Du Q, Li X, Zhang H, Han D. Altered intestinal microbiota enhances adenoid hypertrophy by disrupting the immune balance. Front Immunol 2023; 14:1277351. [PMID: 38090578 PMCID: PMC10715246 DOI: 10.3389/fimmu.2023.1277351] [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: 08/14/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Introduction Adenoid hypertrophy (AH) is a common upper respiratory disorder in children. Disturbances of gut microbiota have been implicated in AH. However, the interplay of alteration of gut microbiome and enlarged adenoids remains elusive. Methods 119 AH children and 100 healthy controls were recruited, and microbiome profiling of fecal samples in participants was performed using 16S rRNA gene sequencing. Fecal microbiome transplantation (FMT) was conducted to verify the effects of gut microbiota on immune response in mice. Results In AH individuals, only a slight decrease of diversity in bacterial community was found, while significant changes of microbial composition were observed between these two groups. Compared with HCs, decreased abundances of Akkermansia, Oscillospiraceae and Eubacterium coprostanoligenes genera and increased abundances of Bacteroides, Faecalibacterium, Ruminococcus gnavus genera were revealed in AH patients. The abundance of Bacteroides remained stable with age in AH children. Notably, a microbial marker panel of 8 OTUs were identified, which discriminated AH from HC individuals with an area under the curve (AUC) of 0.9851 in the discovery set, and verified in the geographically different validation set, achieving an AUC of 0.9782. Furthermore, transfer of mice with fecal microbiota from AH patients dramatically reduced the proportion of Treg subsets within peripheral blood and nasal-associated lymphoid tissue (NALT) and promoted the expansion of Th2 cells in NALT. Conclusion These findings highlight the effect of the altered gut microbiota in the AH pathogenesis.
Collapse
Affiliation(s)
- Wenxin Liu
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huier Jiang
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiling Liu
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Ministry of Justice, Shanghai, China
| | - Yue Zheng
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanan Liu
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fen Pan
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fangyuan Yu
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi Li
- Department of Pathology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Meizhen Gu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qingqing Du
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyan Li
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hong Zhang
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dingding Han
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Medical School, Guangxi University, Nanning, China
| |
Collapse
|
11
|
Alemu BK, Azeze GG, Wu L, Lau SL, Wang CC, Wang Y. Effects of maternal probiotic supplementation on breast milk microbiome and infant gut microbiome and health: a systematic review and meta-analysis of randomized controlled trials. Am J Obstet Gynecol MFM 2023; 5:101148. [PMID: 37660760 DOI: 10.1016/j.ajogmf.2023.101148] [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: 05/30/2023] [Revised: 08/18/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023]
Abstract
OBJECTIVE The early-life microbiome is formed during the perinatal period and is critical for infants' lifelong health. This is established by maternal-infant microbiome crosstalk, which is mediated by the breast milk microbiome. The milk microbiome is dependent on the maternal gut microbiome, suggesting that it could potentially be restored through oral probiotic supplements. Therefore, we conducted this systematic review and meta-analysis to summarize the effect of maternal probiotic supplements on breast milk and infant gut microbiome composition and on infant health. DATA SOURCES The PubMed, EMBASE, Web of Science, Scopus, CINAHL, and Science Direct databases were searched until December 15, 2022. STUDY ELIGIBILITY CRITERIA Randomized controlled trials following the population, intervention, comparison, and outcome (population: pregnant or lactating women; intervention: probiotics; control: placebo or follow-up; outcome: breast milk and infant gut microbiome composition and infant health) principles were included. METHODS Using a random effect model, the standard mean difference, risk difference, and risk ratio with 95% confidence interval were used to measure each outcome. All analyses were conducted using the intention-to-treat approach. Heterogeneity was evaluated using I2 statistics. RESULTS The final data set included 24 randomized controlled trials with a total of 2761 mothers and 1756 infants. The overall effect of probiotics on the beneficial bacteria detection rate in breast milk had a risk difference of 24% (95% confidence interval, 0.1-0.37; P<.001; I2=91.12%). The pooled mean beneficial and pathogenic bacteria abundance in breast milk had a standard mean difference of 1.22 log10 colony forming units/mL (95% confidence interval, 0.48-1.97; P<.001; I2=95.51%) and -1.05 log10 colony forming unites/mL (95% confidence interval, -1.99 to -0.12; P=.03; I2=96.79%), respectively. The overall abundance of beneficial bacteria in the infant gut had a standard mean difference of 0.89 log10 colony forming units/g (95% confidence interval, 0.22-1.56; P=.01; I2=95.01%). It also controlled infant weight gain (standard mean difference, -0.49 kg/equivalent age; 95% confidence interval, -0.82 to -0.17; P<.001; I2=0.00%) and decreased the occurrence of infantile colic (risk ratio, 0.30; 95% confidence interval, 0.16-0.57; P<.001; I2=0.00%). CONCLUSION Maternal probiotic supplements effectively orchestrate the breast milk and infant gut microbiome with a wide range of clinical benefits and safety. Lactobacillus, Bifidobacterium, Streptococcus thermophilus, and S. boulardii can be used as maternal supplements to promote infant health.
Collapse
Affiliation(s)
- Bekalu Kassie Alemu
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR (Messers Alemu and Azeze and Drs Wu, Lau, C Wang, and Y Wang); Department of Midwifery, College of Medicine and Health Sciences, Debre Markos University, Ethiopia (Mr Alemu)
| | - Getnet Gedefaw Azeze
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR (Messers Alemu and Azeze and Drs Wu, Lau, C Wang, and Y Wang); Department of Midwifery, College Medicine and Health Sciences, Injibara University, Ethiopia (Mr Azeze)
| | - Ling Wu
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR (Messers Alemu and Azeze and Drs Wu, Lau, C Wang, and Y Wang)
| | - So Ling Lau
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR (Messers Alemu and Azeze and Drs Wu, Lau, C Wang, and Y Wang)
| | - Chi Chiu Wang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR (Messers Alemu and Azeze and Drs Wu, Lau, C Wang, and Y Wang); Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, Chinese University of Hong Kong-Sichuan University Joint Laboratory for Reproductive Medicine, Hong Kong SAR (Dr C Wang)
| | - Yao Wang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR (Messers Alemu and Azeze and Drs Wu, Lau, C Wang, and Y Wang).
| |
Collapse
|
12
|
Tomaszewska A, Jeleniewska A, Porębska K, Królikowska K, Rustecka A, Lipińska-Opałka A, Będzichowska A, Zdanowski R, Aleksandrowicz K, Kloc M, Kalicki B. Immunomodulatory Effect of Infectious Disease of a Breastfed Child on the Cellular Composition of Breast Milk. Nutrients 2023; 15:3844. [PMID: 37686876 PMCID: PMC10490220 DOI: 10.3390/nu15173844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
Recent studies suggest that the content of immune components in milk is influenced by the mother's health and also by the infant she feeds. We aimed to evaluate the effect of a child's respiratory tract infection on the cellular composition of breast milk (neutrophils, monocytes, eosinophils, lymphocytes, and their subpopulations). Twenty-six breastfeeding mothers whose children were hospitalized for respiratory tract infections were enrolled in the study. The control group consisted of 23 mothers of healthy children. Regarding the children, baseline laboratory blood tests were performed, and nasal swabs were taken for the presence of RS virus. In the next step, milk samples were collected from the mothers to assess the cellular composition of the milk, including neutrophils, monocytes, eosinophils, lymphocytes, and their subpopulations. Significantly higher percentages of T lymphocytes (helper and cytotoxic lymphocytes) were observed in the milk of the studied mothers. There was a significantly higher percentage of milk lymphocytes in the group of affected children with confirmed RSV etiology than in children with excluded RSV etiology. A significant positive correlation was observed between the duration of infection and the percentage of milk NK cells and between milk CD19 lymphocytes and the child's serum leukocytosis. This study may provide evidence of a link between cells in breast milk and disease in the breastfed infant. The severity of the infection, its duration, and the etiological agent of the infection may affect the cellular composition of milk.
Collapse
Affiliation(s)
- Agata Tomaszewska
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| | - Alicja Jeleniewska
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| | - Klaudia Porębska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (K.P.); (R.Z.); (K.A.)
| | - Katarzyna Królikowska
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| | - Agnieszka Rustecka
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| | - Agnieszka Lipińska-Opałka
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| | - Agata Będzichowska
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| | - Robert Zdanowski
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (K.P.); (R.Z.); (K.A.)
| | - Karolina Aleksandrowicz
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (K.P.); (R.Z.); (K.A.)
| | - Małgorzata Kloc
- Transplant Immunology, The Houston Methodist Research Institute, Houston, TX 77030, USA;
- Department of Surgery, The Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Genetics, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA
| | - Bolesław Kalicki
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| |
Collapse
|
13
|
Karimi H, Mansouri V, Rezaei N. Vertical transmission and maternal passive immunity post-SARS-CoV-2. Future Virol 2023:10.2217/fvl-2023-0089. [PMID: 37822684 PMCID: PMC10564388 DOI: 10.2217/fvl-2023-0089] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/22/2023] [Indexed: 10/13/2023]
Abstract
Since 2020, the highly contagious nature and various transmission routes of SARS-CoV-2 have rendered the pandemic interminable. Vertical transmission (VT) through the placenta and breast milk, which is frequent for certain virus types, is thought to exist for SARS-CoV-2 and is hypothesized by many researchers. Conversely, antibodies are produced to counteract the effect of viruses. Since newborns' immunologic system cannot produce proper antibodies, maternal antibodies are usually transferred from mother to infant/fetus to meet the need. This theory leads to the hypothesis of transmission of antibodies through the placenta and breast milk following SARS-CoV-2 infection or vaccination. This paper further discusses these hypotheses, considering consequences of fetus/infant harm versus benefit.
Collapse
Affiliation(s)
- Hanie Karimi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Vahid Mansouri
- Gene Therapy Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, 14194, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Yang X, Fox A, DeCarlo C, Pineda N, Powell RL. The Secretory IgA Response in Human Milk Against the SARS-CoV-2 Spike Is Highly Durable and Neutralizing for At Least 1 Year of Lactation Postinfection. Breastfeed Med 2023; 18:602-611. [PMID: 37615565 PMCID: PMC10460685 DOI: 10.1089/bfm.2023.0117] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Background: Although in the early pandemic period COVID-19 pathology among young children and infants was typically less severe compared with that observed among adults, this has not remained entirely consistent as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants have emerged. There is an enormous body of evidence demonstrating the benefits of human milk antibodies (Abs) in protecting infants against a wide range of enteric and respiratory infections. It is highly plausible that the same holds true for protection against SARS-CoV-2 as this virus infects cells of the gastrointestinal and respiratory mucosae. Understanding the durability of a human milk Ab response over time after infection is critical. Objective: Previously, we examined the Abs present in milk of those recently infected with SARS-CoV-2 and concluded that the response was secretory immunoglobulin A (sIgA) dominant and that these titers were highly correlated with neutralization potency. The present study aimed to monitor the durability of the SARS-CoV-2 IgA and secretory Ab (sAb) response in milk from COVID-19-recovered lactating individuals over 12 months in the absence of vaccination or reinfection. Results: This analysis revealed a robust and durable spike-specific milk sIgA response, and at 9-12 months after infection, 88% of the samples exhibited titers above the positive cutoff for IgA and 94% were above the cutoff for sAb. Fifty percent of participants exhibited less than twofold reduction of spike-specific IgA through 12 months. A strong, significant positive correlation between IgA and sAb against spike persisted throughout the study period. Nucleocapsid-specific Abs were also assessed, which revealed significant background or cross-reactivity of milk IgA against this immunogen, as well as limited/inconsistent durability compared with Spike titers. Conclusion: These data suggest that lactating individuals are likely to continue producing spike-specific Abs in their milk for 1 year or more, which may provide critical passive immunity to infants against SARS-CoV-2 throughout the lactation period.
Collapse
Affiliation(s)
- Xiaoqi Yang
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alisa Fox
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Claire DeCarlo
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nicole Pineda
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rebecca L.R. Powell
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
15
|
Wijenayake S, Martz J, Lapp HE, Storm JA, Champagne FA, Kentner AC. The contributions of parental lactation on offspring development: It's not udder nonsense! Horm Behav 2023; 153:105375. [PMID: 37269591 PMCID: PMC10351876 DOI: 10.1016/j.yhbeh.2023.105375] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 06/05/2023]
Abstract
The Developmental Origins of Health and Disease (DOHaD) hypothesis describes how maternal stress exposures experienced during critical periods of perinatal life are linked to altered developmental trajectories in offspring. Perinatal stress also induces changes in lactogenesis, milk volume, maternal care, and the nutritive and non-nutritive components of milk, affecting short and long-term developmental outcomes in offspring. For instance, selective early life stressors shape the contents of milk, including macro/micronutrients, immune components, microbiota, enzymes, hormones, milk-derived extracellular vesicles, and milk microRNAs. In this review, we highlight the contributions of parental lactation to offspring development by examining changes in the composition of breast milk in response to three well-characterized maternal stressors: nutritive stress, immune stress, and psychological stress. We discuss recent findings in human, animal, and in vitro models, their clinical relevance, study limitations, and potential therapeutic significance to improving human health and infant survival. We also discuss the benefits of enrichment methods and support tools that can be used to improve milk quality and volume as well as related developmental outcomes in offspring. Lastly, we use evidence-based primary literature to convey that even though select maternal stressors may modulate lactation biology (by influencing milk composition) depending on the severity and length of exposure, exclusive and/or prolonged milk feeding may attenuate the negative in utero effects of early life stressors and promote healthy developmental trajectories. Overall, scientific evidence supports lactation to be protective against nutritive and immune stressors, but the benefits of lactation in response to psychological stressors need further investigation.
Collapse
Affiliation(s)
- Sanoji Wijenayake
- Department of Biology, The University of Winnipeg, Winnipeg, Manitoba, Canada.
| | - Julia Martz
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA
| | - Hannah E Lapp
- Deparment of Psychology, University of Texas at Austin, Austin, TX, USA
| | - Jasmyne A Storm
- Department of Biology, The University of Winnipeg, Winnipeg, Manitoba, Canada
| | | | - Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA.
| |
Collapse
|
16
|
Martin‐Vicente M, Carrasco I, Muñoz‐Gomez M, Lobo AH, Mas V, Vigil‐Vázquez S, Vázquez M, Manzanares A, Cano O, Alonso R, Sepúlveda‐Crespo D, Tarancón‐Díez L, Muñoz‐Fernández M, Muñoz‐Chapuli M, Resino S, Navarro ML, Martinez I. Antibody levels to SARS-CoV-2 spike protein in mothers and children from delivery to six months later. Birth 2023; 50:418-427. [PMID: 35802776 PMCID: PMC9349436 DOI: 10.1111/birt.12667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 05/11/2022] [Accepted: 06/15/2022] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Pregnant women are vulnerable to severe acute respiratory syndrome coronavirus (SARS-CoV-2) infection. Neutralizing antibodies against the SARS-CoV-2 spike (S) protein protect from severe disease. This study analyzes the antibody titers to SARS-CoV-2 S protein in pregnant women and their newborns at delivery, and six months later. METHODS We conducted a prospective study on pregnant women with confirmed SARS-CoV-2 infection and newborns. Antibody (IgG, IgM, and IgA) titers were determined using immunoassays in serum and milk samples. An angiotensin-converting enzyme 2 (ACE2) receptor-binding inhibition assay to the S protein was performed on the same serum and milk samples. RESULTS At birth, antibodies to SARS-CoV-2 spike protein were detected in 81.9% of mothers' sera, 78.9% of cord blood samples, and 63.2% of milk samples. Symptomatic women had higher antibody titers (IgG, IgM, and IgA) than the asymptomatic ones (P < 0.05). At six months postpartum, IgG levels decreased drastically in children's serum (P < 0.001) but remained high in mothers' serum. Antibody titers correlated positively with its capacity to inhibit the ACE2-spike protein interaction at baseline in maternal sera (R2 = 0.203; P < 0.001), cord sera (R2 = 0.378; P < 0.001), and milk (R2 = 0.564; P < 0.001), and at six months in maternal sera (R2 = 0.600; P < 0.001). CONCLUSIONS High antibody levels against SARS-CoV-2 spike protein were found in most pregnant women. Due to the efficient transfer of IgG to cord blood and high IgA titers in breast milk, neonates may be passively immunized to SARS-CoV-2 infection. Our findings could guide newborn management and maternal vaccination policies.
Collapse
Affiliation(s)
- María Martin‐Vicente
- Unidad de Infección Viral e InmunidadCentro Nacional de Microbiología, Instituto de Salud Carlos III, MajadahondaMadridSpain
| | - Itziar Carrasco
- Grupo de Investigación en Infectología PediátricaInstituto de Investigación Sanitaria Gregorio MarañónMadridSpain
| | - María José Muñoz‐Gomez
- Unidad de Infección Viral e InmunidadCentro Nacional de Microbiología, Instituto de Salud Carlos III, MajadahondaMadridSpain
| | - Alicia Hernanz Lobo
- Grupo de Investigación en Infectología PediátricaInstituto de Investigación Sanitaria Gregorio MarañónMadridSpain
- Servicio de PediatríaHospital General Universitario Gregorio MarañónMadridSpain
| | - Vicente Mas
- Unidad de Biología ViralCentro Nacional de Microbiología, Instituto de Salud Carlos IIIMadridSpain
| | - Sara Vigil‐Vázquez
- Sevicio de NeonatologíaHospital General Universitario Gregorio MarañónMadridSpain
| | - Mónica Vázquez
- Unidad de Biología ViralCentro Nacional de Microbiología, Instituto de Salud Carlos IIIMadridSpain
| | - Angela Manzanares
- Servicio de PediatríaHospital General Universitario Gregorio MarañónMadridSpain
| | - Olga Cano
- Unidad de Biología ViralCentro Nacional de Microbiología, Instituto de Salud Carlos IIIMadridSpain
| | - Roberto Alonso
- Departamento de Microbiología Clínica y Enfermedades InfecciosasHospital General Universitario Gregorio MarañónMadridSpain
| | - Daniel Sepúlveda‐Crespo
- Unidad de Infección Viral e InmunidadCentro Nacional de Microbiología, Instituto de Salud Carlos III, MajadahondaMadridSpain
| | - Laura Tarancón‐Díez
- Laboratorio de InmunoBiología Molecular, Sección de InmunologíaHospital General Universitario Gregorio MarañónMadridSpain
| | - María Ángeles Muñoz‐Fernández
- Laboratorio de InmunoBiología Molecular, Sección de InmunologíaHospital General Universitario Gregorio MarañónMadridSpain
- HIV‐HGM BioBankMadridSpain
| | - Mar Muñoz‐Chapuli
- Departamento de Obstetricia y GinecologíaHospital General Universitario Gregorio MarañónMadridSpain
| | - Salvador Resino
- Unidad de Infección Viral e InmunidadCentro Nacional de Microbiología, Instituto de Salud Carlos III, MajadahondaMadridSpain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC)Instituto de Salud Carlos IIIMadridSpain
| | - Maria Luisa Navarro
- Grupo de Investigación en Infectología PediátricaInstituto de Investigación Sanitaria Gregorio MarañónMadridSpain
- Servicio de PediatríaHospital General Universitario Gregorio MarañónMadridSpain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC)Instituto de Salud Carlos IIIMadridSpain
| | - Isidoro Martinez
- Unidad de Infección Viral e InmunidadCentro Nacional de Microbiología, Instituto de Salud Carlos III, MajadahondaMadridSpain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC)Instituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
17
|
Yang X, DeCarlo C, Fox A, Pineda N, Powell RLR. Assessment of human milk samples obtained pre and post-influenza vaccination reveals a poor boosting of seasonally-relevant, hemagglutinin-specific antibodies. Front Immunol 2023; 14:1154782. [PMID: 37325620 PMCID: PMC10264617 DOI: 10.3389/fimmu.2023.1154782] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction Influenza (flu) vaccination prevented over 100,000 hospitalizations and 7000 deaths from flu over the 2019-2020 season in the USA. Infants <6 months are the most likely to die from flu, though flu vaccines are only licensed for infants >6 months old. Therefore, it is recommended that flu vaccination occur during pregnancy, as this reduces severe complications; however, vaccination rates are suboptimal, and vaccination is also recommended postpartum. For breast/chest-fed infants, the vaccine is believed to elicit protective and robust seasonally-specific milk antibody (Ab). Few comprehensive studies exist examining Ab responses in milk after vaccination, with none measuring secretory Ab (sAb). Determining whether sAbs are elicited is critical, as this Ab class is highly stable in milk and mucosae. Methods In the present study, our aim was to determine to what extent specific Ab titers in the milk of lactating people were boosted after seasonal influenza vaccination. Over the 2019-2020 and 2020-2021 seasons, milk was obtained pre- and post-vaccination and assessed for specific IgA, IgG, and sAb against relevant hemagglutinin (HA) antigens by a Luminex immunoassay. Results IgA and sAb were not found to be significantly boosted, while only IgG titers against B/Phuket/3073/2013, included in vaccines since 2015, exhibited an increase. Across the 7 immunogens examined, as many as 54% of samples exhibited no sAb boost. No significant differences for IgA, sAb, or IgG boosting were measured between seasonally-matched versus mismatched milk groups, indicating boosting was not seasonally-specific. No correlations between IgA and sAb increases were found for 6/8 HA antigens. No boost in IgG- or IgA-mediated neutralization post vaccination was observed. Discussion This study highlights the critical need to redesign influenza vaccines with the lactating population in mind, wherein the aim should be to elicit a potent seasonally-specific sAb response in milk. As such, this population must be included in clinical studies.
Collapse
Affiliation(s)
| | | | | | | | - Rebecca L. R. Powell
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
18
|
Yang X, Fox A, DeCarlo C, Pineda N, Powell RL. The secretory IgA (sIgA) response in human milk against the SARS-CoV-2 Spike is highly durable and neutralizing for at least 1 year of lactation post-infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.19.23290192. [PMID: 37293109 PMCID: PMC10246141 DOI: 10.1101/2023.05.19.23290192] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Although in the early pandemic period, COVID-19 pathology among young children and infants was typically less severe compared to that observed among adults, this has not remained entirely consistent as SARS-CoV-2 variants have emerged. There is an enormous body of evidence demonstrating the benefits of human milk antibodies (Abs) in protecting infants against a wide range of enteric and respiratory infections. It is highly plausible that the same holds true for protection against SARS-CoV-2, as this virus infects cells of the gastrointestinal and respiratory mucosae. Understanding the durability of a human milk Ab response over time after infection is critical. Previously, we examined the Abs present in milk of those recently infected with SARS-CoV-2, and concluded that the response was secretory IgA (sIgA)-dominant and that these titers were highly correlated with neutralization potency. The present study aimed to monitor the durability of the SARS-CoV-2 IgA and secretory Ab (sAb) response in milk from COVID-19-recovered lactating individuals over 12 months, in the absence of vaccination or re-infection. This analysis revealed a robust and durable Spike-specific milk sIgA response, that at 9-12 months after infection, 88% of the samples exhibited titers above the positive cutoff for IgA and 94% were above cutoff for sAb. Fifty percent of participants exhibited less than a 2-fold reduction of Spike-specific IgA through 12 months. A strong significant positive correlation between IgA and sAb against Spike persisted throughout the study period. Nucleocapsid-specific Abs were also assessed, which revealed significant background or cross reactivity of milk IgA against this immunogen, as well as limited/inconsistent durability compared to Spike titers. These data suggests that lactating individuals are likely to continue producing Spike-specific Abs in their milk for 1 year or more, which may provide critical passive immunity to infants against SARS-CoV-2 throughout the lactation period.
Collapse
Affiliation(s)
- Xiaoqi Yang
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York, USA
| | - Alisa Fox
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York, USA
| | - Claire DeCarlo
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York, USA
| | - Nicole Pineda
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York, USA
| | - Rebecca L.R. Powell
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York, USA
| |
Collapse
|
19
|
LeMaster C, Pierce SH, Geanes ES, Khanal S, Elliott SS, Scott AB, Louiselle DA, McLennan R, Maulik D, Lewis T, Pastinen T, Bradley T. The cellular and immunological dynamics of early and transitional human milk. Commun Biol 2023; 6:539. [PMID: 37202439 DOI: 10.1038/s42003-023-04910-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 05/03/2023] [Indexed: 05/20/2023] Open
Abstract
Human milk is essential for infant nutrition and immunity, providing protection against infections and other immune-mediated diseases during the lactation period and beyond in later childhood. Milk contains a broad range of bioactive factors such as nutrients, hormones, enzymes, immunoglobulins, growth factors, cytokines, and antimicrobial factors, as well as heterogeneous populations of maternal cells. The soluble and cellular components of milk are dynamic over time to meet the needs of the growing infant. In this study, we utilize systems-approaches to define and characterize 62 analytes of the soluble component, including immunoglobulin isotypes, as well as the cellular component of human milk during the first two weeks postpartum from 36 mothers. We identify soluble immune and growth factors that are dynamic over time and could be utilized to classify milk into different phenotypic groups. We identify 24 distinct populations of both epithelial and immune cells by single-cell transcriptome analysis of 128,016 human milk cells. We found that macrophage populations have shifting inflammatory profiles during the first two weeks of lactation. This analysis provides key insights into the soluble and cellular components of human milk and serves as a substantial resource for future studies of human milk.
Collapse
Affiliation(s)
- Cas LeMaster
- Genomic Medicine Center, Children's Mercy Research Institute, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Stephen H Pierce
- Genomic Medicine Center, Children's Mercy Research Institute, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Eric S Geanes
- Genomic Medicine Center, Children's Mercy Research Institute, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Santosh Khanal
- Genomic Medicine Center, Children's Mercy Research Institute, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Staci S Elliott
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Allison B Scott
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Daniel A Louiselle
- Genomic Medicine Center, Children's Mercy Research Institute, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Rebecca McLennan
- Genomic Medicine Center, Children's Mercy Research Institute, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Devika Maulik
- Fetal Health Center, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Tamorah Lewis
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- Department of Pediatrics, UMKC School of Medicine, Kansas City, MO, 64108, USA
| | - Tomi Pastinen
- Genomic Medicine Center, Children's Mercy Research Institute, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- Department of Pediatrics, UMKC School of Medicine, Kansas City, MO, 64108, USA
| | - Todd Bradley
- Genomic Medicine Center, Children's Mercy Research Institute, Children's Mercy Kansas City, Kansas City, MO, 64108, USA.
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
- Department of Pediatrics, UMKC School of Medicine, Kansas City, MO, 64108, USA.
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
20
|
Tanaka M, Date M, Miura K, Ito M, Mizuno N, Mizuno K. Protein and Immune Component Content of Donor Human Milk in Japan: Variation with Gestational and Postpartum Age. Nutrients 2023; 15:2278. [PMID: 37242161 PMCID: PMC10221383 DOI: 10.3390/nu15102278] [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: 04/10/2023] [Revised: 05/02/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Donor human milk (DHM) is the second-best nutrition for preterm infants when their own mother's milk is unavailable. The nutrient content of human milk is influenced by various factors, including gestational and postpartum age, but there are no data regarding DHM composition in Japan. The aim of this study was to determine the protein and immune component content of DHM in Japan and to elucidate the effects of gestational and postpartum age on nutrient composition. From September 2021 to May 2022, 134 DHM samples were collected from 92 mothers of preterm and term infants. Protein concentrations in preterm DHM (n = 41) and term DHM (n = 93) were analyzed using a Miris Human Milk Analyzer. The concentrations of secretory immunoglobulin A (sIgA) and lactoferrin, major immune components, were measured using enzyme-linked immunosorbent assays. Preterm DHM exhibited higher protein content than term DHM (1.2 g/dL and 1.0 g/dL, respectively, p < 0.001), whereas sIgA content was higher in term DHM than in preterm DHM (110 μg/mL and 68.4 μg/mL, respectively, p < 0.001). Gestational age was negatively correlated with protein levels and positively correlated with sIgA and lactoferrin levels. Furthermore, a negative correlation was found between postpartum week and protein, sIgA, and lactoferrin concentrations. Our data suggest that gestational and postpartum age affects protein, sIgA, and lactoferrin concentrations in DHM. These results indicate the importance of nutritional analysis for the appropriate use of DHM in preterm infants.
Collapse
Affiliation(s)
- Miori Tanaka
- The Nippon Foundation Human Milk Bank, 17-10 Nihonbashi-koamicho, Chuo-ku, Tokyo 103-0016, Japan; (M.D.); (K.M.); (M.I.); (K.M.)
| | - Midori Date
- The Nippon Foundation Human Milk Bank, 17-10 Nihonbashi-koamicho, Chuo-ku, Tokyo 103-0016, Japan; (M.D.); (K.M.); (M.I.); (K.M.)
| | - Kumiko Miura
- The Nippon Foundation Human Milk Bank, 17-10 Nihonbashi-koamicho, Chuo-ku, Tokyo 103-0016, Japan; (M.D.); (K.M.); (M.I.); (K.M.)
- Faculty of Medicine, Oita University, 1-1 Hasamamachiidaigaoka, Yufu-shi, Oita 879-5593, Japan
| | - Mizuho Ito
- The Nippon Foundation Human Milk Bank, 17-10 Nihonbashi-koamicho, Chuo-ku, Tokyo 103-0016, Japan; (M.D.); (K.M.); (M.I.); (K.M.)
- School of Medicine, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Noriko Mizuno
- Japan Human Milk Bank Association, 4-4 Nihonbashi-hisamatsucho, Chuo-ku, Tokyo 103-8480, Japan;
| | - Katsumi Mizuno
- The Nippon Foundation Human Milk Bank, 17-10 Nihonbashi-koamicho, Chuo-ku, Tokyo 103-0016, Japan; (M.D.); (K.M.); (M.I.); (K.M.)
- Japan Human Milk Bank Association, 4-4 Nihonbashi-hisamatsucho, Chuo-ku, Tokyo 103-8480, Japan;
- Department of Pediatrics, School of Medicine, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| |
Collapse
|
21
|
Mo K, Yu W, Li J, Zhang Y, Xu Y, Huang X, Ni H. Dietary supplementation with a microencapsulated complex of thymol, carvacrol, and cinnamaldehyde improves intestinal barrier function in weaning piglets. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:1994-2003. [PMID: 36347590 DOI: 10.1002/jsfa.12322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/19/2022] [Accepted: 11/09/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND The authors previously prepared a microencapsulated complex of thymol, carvacrol, and cinnamaldehyde (MEEO). This study aimed to evaluate the effect of MEEO on the intestinal mucosal barrier and homeostasis in weaning piglets. A comparison of the effect of MEEO versus chlortetracycline (CTC) was performed in this study. RESULTS Piglets were divided into three groups - control (Con), MEEO, and CTC groups - and raised for 28 days. The results showed that MEEO significantly elevated the ratio of the villus height and the crypt depth in the jejunum and decreased the crypt depth in the ileum compared with the other groups (P < 0.05); it also upregulated the messenger ribonucleic acid (mRNA) expression of tight junction protein in the small intestine. Compared with the Con group, MEEO increased the concentration of secretory immunoglobulin A (sIgA), cathelicidin antimicrobial peptides (CAMP), and interleukin 10 (IL-10), while decreasing the interleukin 1 beta (IL-1β) concentration in both jejunal and ileal mucosa (P < 0.05). The mRNA expression of jejunal mucosal MUC1 and ileal mucosal MUC2 was increased in the MEEO group compared with the other groups (P < 0.05). Intestinal microbial analysis showed that dietary treatment had little impact on the ileal microbial structure. A significant rise in the genus Lactobacillus was, however, found in the MEEO group. There is a positive correlation between the Lactobacillus and sIgA, and between the Lactobacillus and CAMP, indicating that an improvement in the mucosal barrier function by the addition of MEEO may be associated with the proliferation of Lactobacillus. CONCLUSION Dietary supplementation with MEEO improves intestinal barrier function in weaning piglets, the effect of which was superior to CTC. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Kaibin Mo
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Wengang Yu
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jing Li
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Yunxiao Zhang
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ying Xu
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xianhui Huang
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Hengjia Ni
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| |
Collapse
|
22
|
Li Y, Wang J, Liu Q, Yang F, Chen L, Xu H, Jin W, Liu Y, Zhang W, Yang F, He Z, Zhao Y. Composition of the intestinal microbiota of infant rhesus macaques at different ages before and after weaning. Heliyon 2023; 9:e13915. [PMID: 36923844 PMCID: PMC10009685 DOI: 10.1016/j.heliyon.2023.e13915] [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: 08/16/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
Background Rhesus macaques and humans are closely related genetically and share similar physiological and pathological characteristics. Exploring the impact of diet on the early establishment of gut microbiota in non-human primates can provide relevant clinical models for healthy infant growth and development. At present, few writers have focused on the composition and changes of the intestinal microbes of infant rhesus macaques throughout their progression from birth to formula feeding after weaning. In this study, we used 16S rRNA sequencing technology to explore the composition of the intestinal flora of rhesus macaques at different ages and analyzed the trends in the microbial changes. Results The results showed that the relative abundance of Bifidobacterium and Lactobacillus in the intestinal flora of infant rhesus macaques significantly decreased, and Prevotella increased with age. Bifidobacterium longum and Bifidobacterium breve are effective biomarkers to predict grouping. The metabolic pathways enriched in early life mainly concentrated in glycosphingolipid biosynthesis (lacto and neolacto series) and the degradation and metabolism of alcohols and esters. Conclusions We found that age was an important factor that affected the changes in the intestinal flora. This study revealed the change trend of flora in breastfed and formula-fed infant rhesus monkeys in different growth months, and found that the dominant flora changed greatly. This research provides a medically relevant theoretical basis for understanding the healthy development of infants.
Collapse
Affiliation(s)
- Yanyan Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China.,Yunnan Key Laboratory of Major Infections Diseases Vaccine Development, Kunming, Yunnan Province, 650118, China
| | - Junbin Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China.,Yunnan Key Laboratory of Major Infections Diseases Vaccine Development, Kunming, Yunnan Province, 650118, China
| | - Quan Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China.,Yunnan Key Laboratory of Major Infections Diseases Vaccine Development, Kunming, Yunnan Province, 650118, China
| | - Fengmei Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China.,Yunnan Key Laboratory of Major Infections Diseases Vaccine Development, Kunming, Yunnan Province, 650118, China
| | - Lixiong Chen
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China.,Yunnan Key Laboratory of Major Infections Diseases Vaccine Development, Kunming, Yunnan Province, 650118, China
| | - Hongjie Xu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China.,Yunnan Key Laboratory of Major Infections Diseases Vaccine Development, Kunming, Yunnan Province, 650118, China
| | - Weihua Jin
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China.,Yunnan Key Laboratory of Major Infections Diseases Vaccine Development, Kunming, Yunnan Province, 650118, China
| | - Yu Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China.,Yunnan Key Laboratory of Major Infections Diseases Vaccine Development, Kunming, Yunnan Province, 650118, China
| | - Wei Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China.,Yunnan Key Laboratory of Major Infections Diseases Vaccine Development, Kunming, Yunnan Province, 650118, China
| | - Fei Yang
- Department of Thoracic Surgery, Kunming Third People's Hospital, Kunming, Yunnan Province, 650041, China
| | - Zhanlong He
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China.,Yunnan Key Laboratory of Major Infections Diseases Vaccine Development, Kunming, Yunnan Province, 650118, China
| | - Yuan Zhao
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China.,Yunnan Key Laboratory of Major Infections Diseases Vaccine Development, Kunming, Yunnan Province, 650118, China
| |
Collapse
|
23
|
Chen YY, Tun HM, Field CJ, Mandhane PJ, Moraes TJ, Simons E, Turvey SE, Subbarao P, Scott JA, Kozyrskyj AL. Impact of Cesarean Delivery and Breastfeeding on Secretory Immunoglobulin A in the Infant Gut Is Mediated by Gut Microbiota and Metabolites. Metabolites 2023; 13:metabo13020148. [PMID: 36837767 PMCID: PMC9959734 DOI: 10.3390/metabo13020148] [Citation(s) in RCA: 2] [Impact Index Per Article: 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
|
24
|
Ogony J, Hoskin TL, Stallings-Mann M, Winham S, Brahmbhatt R, Arshad MA, Kannan N, Peña A, Allers T, Brown A, Sherman ME, Visscher DW, Knutson KL, Radisky DC, Degnim AC. Immune cells are increased in normal breast tissues of BRCA1/2 mutation carriers. Breast Cancer Res Treat 2023; 197:277-285. [PMID: 36380012 PMCID: PMC10168666 DOI: 10.1007/s10549-022-06786-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/25/2022] [Indexed: 11/17/2022]
Abstract
PURPOSE Breast cancer risk is elevated in pathogenic germline BRCA 1/2 mutation carriers due to compromised DNA quality control. We hypothesized that if immunosurveillance promotes tumor suppression, then normal/benign breast lobules from BRCA carriers may demonstrate higher immune cell densities. METHODS We assessed immune cell composition in normal/benign breast lobules from age-matched women with progressively increased breast cancer risk, including (1) low risk: 19 women who donated normal breast tissue to the Komen Tissue Bank (KTB) at Indiana University Simon Cancer Center, (2) intermediate risk: 15 women with biopsy-identified benign breast disease (BBD), and (3) high risk: 19 prophylactic mastectomies from women with germline mutations in BRCA1/2 genes. We performed immunohistochemical stains and analysis to quantitate immune cell densities from digital images in up to 10 representative lobules per sample. Median cell counts per mm2 were compared between groups using Wilcoxon rank-sum tests. RESULTS Normal/benign breast lobules from BRCA carriers had significantly higher densities of immune cells/mm2 compared to KTB normal donors (all p < 0.001): CD8 + 354.4 vs 150.9; CD4 + 116.3 vs 17.7; CD68 + 237.5 vs 57.8; and CD11c + (3.5% vs 0.4% pixels positive). BBD tissues differed from BRCA carriers only in CD8 + cells but had higher densities of CD4 + , CD11c + , and CD68 + immune cells compared to KTB donors. CONCLUSIONS These preliminary analyses show that normal/benign breast lobules of BRCA mutation carriers contain increased immune cells compared with normal donor breast tissues, and BBD tissues appear overall more similar to BRCA carriers.
Collapse
Affiliation(s)
- Joshua Ogony
- Quantitative Health Sciences, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Tanya L Hoskin
- Quantitative Health Sciences, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Melody Stallings-Mann
- Department of Cancer Biology, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Stacey Winham
- Quantitative Health Sciences, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Rushin Brahmbhatt
- Department of Surgery, Mayo Clinic College of Medicine, 200 First St SW, Rochester, MN, 55905, USA
| | - Muhammad Asad Arshad
- Department of Surgery, Mayo Clinic College of Medicine, 200 First St SW, Rochester, MN, 55905, USA
| | - Nagarajan Kannan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Alvaro Peña
- Department of Surgery, Mayo Clinic College of Medicine, 200 First St SW, Rochester, MN, 55905, USA
| | - Teresa Allers
- Department of Surgery, Mayo Clinic College of Medicine, 200 First St SW, Rochester, MN, 55905, USA
| | - Alyssa Brown
- Mayo Graduate School, Mayo Clinic, Rochester, MN, USA
| | - Mark E Sherman
- Quantitative Health Sciences, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Daniel W Visscher
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Amy C Degnim
- Department of Surgery, Mayo Clinic College of Medicine, 200 First St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
25
|
Saha P, Mell B, Golonka RM, Bovilla VR, Abokor AA, Mei X, Yeoh BS, Doris PA, Gewirtz AT, Joe B, Vijay-Kumar M. Selective IgA Deficiency in Spontaneously Hypertensive Rats With Gut Dysbiosis. Hypertension 2022; 79:2239-2249. [PMID: 35950503 PMCID: PMC9458624 DOI: 10.1161/hypertensionaha.122.19307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/07/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND The spontaneously hypertensive rat (SHR) is extensively used to study hypertension. Gut microbiota dysbiosis is a notable feature in SHR for reasons unknown. Immunoglobulin A (IgA) is a major host factor required for gut microbiota homeostasis. We hypothesized that inadequate IgA contributes to gut microbiota dysbiosis in SHR. METHODS IgA was measured in feces, cecum, serum, liver, gut-associated lymphoid tissue, and milk from SHR and Wistar Kyoto rats. IgA regulatory factors like IgM, IgG, and pIgR (polymeric immunoglobulin receptor) were analyzed. IgA and IgG antibodies and blood pressure (BP) were measured before and after administrating a bacterial antigen (ie, flagellin). RESULTS Compared with Wistar Kyoto rats, SHR displayed remarkably near-deficient IgA levels accompanied by compensatory increases in serum IgM and IgG and gut-liver pIgR expression. Inadequate milk IgA in SHR emphasized this immune defect stemmed from the neonatal stage. Reduced IgA+ B cells in circulation and Peyer patches indicated a possible reason for the lower IgA in SHR. Noteworthy, a genetic insufficiency was unlikely because administering flagellin to SHR induced anti-flagellin IgA antibodies. This immune response surprisingly accelerated hypertension development in SHR, suggesting IgA quiescence may help maintain lower BP. CONCLUSIONS This study is the first to reveal IgA deficiency in SHR as one host factor associated with gut microbiota dysbiosis and invigorates future research to determine the pathophysiological role of IgA in hypertension.
Collapse
Affiliation(s)
- Piu Saha
- UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Blair Mell
- UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Rachel M. Golonka
- UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Venugopal R. Bovilla
- UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Ahmed A. Abokor
- UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Xue Mei
- UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Beng San Yeoh
- UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Peter A. Doris
- Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX 77030, USA
| | - Andrew T. Gewirtz
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Bina Joe
- UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Matam Vijay-Kumar
- UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| |
Collapse
|
26
|
Calabretto M, D'Alisa R, Faraone S, Mazzuti L, Pecorini F, Turriziani O. Detection of SARS-CoV-2 RNA and antibodies in breast milk of infected mothers. J Med Virol 2022; 95:e28142. [PMID: 36098372 PMCID: PMC9538366 DOI: 10.1002/jmv.28142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/19/2022] [Accepted: 09/09/2022] [Indexed: 01/11/2023]
Affiliation(s)
| | - Rossella D'Alisa
- Department of Maternal Infantile and Urological SciencesSapienza University of RomeRomeItaly
| | - Sara Faraone
- Department of Molecular MedicineSapienza University of RomeRomeItaly
| | - Laura Mazzuti
- Department of Molecular MedicineSapienza University of RomeRomeItaly
| | - Francesco Pecorini
- Department of Maternal Infantile and Urological SciencesSapienza University of RomeRomeItaly
| | | |
Collapse
|
27
|
Narayanaswamy V, Pentecost BT, Telfer JC, Burnside AS, Schneider SS, Alfandari D, Baker RL, Saiju A, Nodiff S, Arcaro KF. Durable antibody and effector memory T cell responses in breastmilk from women with SARS-CoV-2. Front Immunol 2022; 13:985226. [PMID: 36172379 PMCID: PMC9512087 DOI: 10.3389/fimmu.2022.985226] [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: 07/03/2022] [Accepted: 08/24/2022] [Indexed: 11/24/2022] Open
Abstract
Background Given that only 25% of pregnant women elect to receive a COVID-19 vaccine, maternal SARS-CoV-2 infection remains an important route of conferring protective passive immunity to breastfed infants of mothers who are not vaccinated. Methods We enrolled 30 lactating participants between December 2020 and March 2021 who had a positive PCR-test and their first COVID-19 symptoms within the previous 21 days. Participants were asked to provide serial bilateral milk samples at 12 timepoints (~ every 3 days) over a period of 35 days. A second set of samples was collected at least four months after the beginning of the first set. Participants also were asked to provide their dried blood spots and infant stool samples. All samples were tested for receptor-binding domain (RBD)-specific immunoglobulin (Ig)A, IgG, and IgM. Milk samples were assessed for neutralizing ability against the spike protein and four SARS-CoV-2 variants: D614G, Alpha (B.1.1.7), Beta (B.1.351), and Gamma (P.1). Permeability of the breast epithelium was assessed by measuring the sodium to potassium ions (Na:K) in milk. Using flow cytometry, memory CD4 and CD8 T cells (CD45RO+ and CCR7+/-) and mucosal-homing CD4 and CD8 T cells (CD103+) were determined in cells from milk expressed at 35 days and at least 4 months after their first milk donation. Results Milk antibodies from SARS-CoV-2 positive participants neutralized the spike complex. Milk from 73, 90, and 53% of participants had binding reactivities to RBD-specific IgA, IgG, and IgM, respectively. In contrast to blood spots, which showed increased levels of IgG, but not IgA or IgM, the COVID-19 response in milk was associated with a robust IgA response. Twenty-seven percent of participants had increased breast-epithelium permeability, as indicated by Na:K ≥ 0.6. The percentage of CD45RO+CCR7- effector-memory T cells in the day ≥120 milk samples was significantly higher than day 35 samples (P< 0.05). Conclusions Antibodies in milk from participants with recent SARS-CoV-2 infection and those who recovered can neutralize the spike complex. For the first time we show that breastmilk T cells are enriched for mucosal memory T cells, further emphasizing the passive protection against SARS-CoV-2 conferred to infants via breastmilk.
Collapse
Affiliation(s)
- Vignesh Narayanaswamy
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Brian T. Pentecost
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Janice C. Telfer
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Amy S. Burnside
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Sallie S. Schneider
- Pioneer Valley Life Sciences Institute, Baystate Medical Center, Springfield, MA, United States
| | - Dominique Alfandari
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Ryan L. Baker
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Aman Saiju
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Sam Nodiff
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Kathleen F. Arcaro
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| |
Collapse
|
28
|
Comparative Proteomic Analysis of Proteins in Breast Milk during Different Lactation Periods. Nutrients 2022; 14:nu14173648. [PMID: 36079904 PMCID: PMC9460426 DOI: 10.3390/nu14173648] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 08/30/2022] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
Breast milk is an unparalleled food for infants, as it can meet almost all of their nutritional needs. Breast milk in the first month is an important source of acquired immunity. However, breast milk protein may vary with the stage of lactation. Therefore, the aim of this study was to use a data-independent acquisition approach to determine the differences in the proteins of breast milk during different lactation periods. The study samples were colostrum (3-6 days), transitional milk (7-14 days), and mature milk (15-29 days). The results identified a total of 2085 different proteins, and colostrum contained the most characteristic proteins. Protein expression was affected by the lactation stage. The proteins expressed in breast milk changed greatly between day 3 and day 14 and gradually stabilized after 14 days. The expression levels of lactoferrin, immunoglobulin, and clusterin were the highest in colostrum. CTP synthase 1, C-type lectin domain family 19 member A, secretoglobin family 3A member 2, trefoil factor 3 (TFF3), and tenascin were also the highest in colostrum. This study provides further insights into the protein composition of breast milk and the necessary support for the design and production of infant formula.
Collapse
|
29
|
Davis EC, Castagna VP, Sela DA, Hillard MA, Lindberg S, Mantis NJ, Seppo AE, Järvinen KM. Gut microbiome and breast-feeding: Implications for early immune development. J Allergy Clin Immunol 2022; 150:523-534. [PMID: 36075638 PMCID: PMC9463492 DOI: 10.1016/j.jaci.2022.07.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022]
Abstract
Establishment of the gut microbiome during early life is a complex process with lasting implications for an individual's health. Several factors influence microbial assembly; however, breast-feeding is recognized as one of the most influential drivers of gut microbiome composition during infancy, with potential implications for function. Differences in gut microbial communities between breast-fed and formula-fed infants have been consistently observed and are hypothesized to partially mediate the relationships between breast-feeding and decreased risk for numerous communicable and noncommunicable diseases in early life. Despite decades of research on the gut microbiome of breast-fed infants, there are large scientific gaps in understanding how human milk has evolved to support microbial and immune development. This review will summarize the evidence on how breast-feeding broadly affects the composition and function of the early-life gut microbiome and discuss mechanisms by which specific human milk components shape intestinal bacterial colonization, succession, and function.
Collapse
Affiliation(s)
- Erin C Davis
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY
| | | | - David A Sela
- Department of Food Science, University of Massachusetts Amherst, Amherst, Mass; Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Mass; Organismic and Evolutionary Biology Graduate Program, University of Massachusetts Amherst, Amherst, Mass
| | - Margaret A Hillard
- Department of Food Science, University of Massachusetts Amherst, Amherst, Mass; Organismic and Evolutionary Biology Graduate Program, University of Massachusetts Amherst, Amherst, Mass
| | - Samantha Lindberg
- Department of Biomedical Sciences, University of Albany, Rensselaer, NY
| | - Nicholas J Mantis
- Division of Infectious Diseases, New York State Department of Health, Albany, NY
| | - Antti E Seppo
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY
| | - Kirsi M Järvinen
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY; Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY; Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY.
| |
Collapse
|
30
|
Szczygioł P, Łukianowski B, Kościelska-Kasprzak K, Jakuszko K, Bartoszek D, Krajewska M, Królak-Olejnik B. Antibodies in the breastmilk of COVID-19 recovered women. BMC Pregnancy Childbirth 2022; 22:635. [PMID: 35953773 PMCID: PMC9368700 DOI: 10.1186/s12884-022-04945-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/25/2022] [Indexed: 11/24/2022] Open
Abstract
Objective Human milk contains antibodies against Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) which may serve as a protective factor through passive immunization in infants. The objective of this study was to measure the levels of anti-SARS-CoV-2 IgG and IgA in human milk and serum after a SARS-CoV-2 infection. Design Breast milk and serum samples from 72 lactating mothers with confirmed SARS-CoV-2 asymptomatic or symptomatic infection were collected 1-229 days after the onset of clinical symptoms related to COVID-19. Seventeen mothers with no history of COVID-19 served as a control group. Enzyme-Linked ImmunoSorbent Assay was performed to analyze antibodies against SARS-CoV-2. Results SARS-CoV-2-IgA human milk antibodies were detected in mothers and their concentrations were consistently higher than SARS-CoV-2-IgG antibodies. The serum and breastmilk samples of women with COVID-19 was characterized by a higher concentration of anti-RBD IgA and IgG than the serum from the control group without COVID-19. No statistically significant difference was observed between the antibody levels in the serum samples obtained from symptomatic and asymptomatic women exposed to SARS-CoV-2 and between the antibody level and the time from a positive SARS-CoV-2 test result over the period studied. Conclusion Our results confirm the presence of SARS-CoV-2 IgA and IgG antibodies in the breastmilk of COVID-19 recovered women and the possibility of these antibodies in providing specific immunologic benefits to breastfeeding infants such as protection against the virus transmission and severity of the acquired COVID-19 disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12884-022-04945-z.
Collapse
Affiliation(s)
- Paulina Szczygioł
- Department of Neonatology, Wroclaw Medical University, 50-367, Wroclaw, Poland.
| | - Błażej Łukianowski
- Department of Neonatology, Wroclaw Medical University, 50-367, Wroclaw, Poland
| | | | - Katarzyna Jakuszko
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, 50-367, Wroclaw, Poland
| | - Dorota Bartoszek
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, 50-367, Wroclaw, Poland
| | - Magdalena Krajewska
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, 50-367, Wroclaw, Poland
| | | |
Collapse
|
31
|
Zheng Y, Correa-Silva S, Palmeira P, Carneiro-Sampaio M. Maternal vaccination as an additional approach to improve the protection of the nursling: Anti-infective properties of breast milk. Clinics (Sao Paulo) 2022; 77:100093. [PMID: 35963149 PMCID: PMC9382412 DOI: 10.1016/j.clinsp.2022.100093] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/08/2022] [Accepted: 07/14/2022] [Indexed: 11/30/2022] Open
Abstract
Human milk constitutes a secretion with unique functions of both nourishing the nursling and providing protection against enteric and respiratory infections, mainly due to its content of secretory IgA antibodies but also due to the presence of a plethora of bioactive factors. Specific IgA antibodies are produced locally by plasma cells derived from B lymphocytes that migrate from other mucosae to the mammary gland during lactation, particularly from the gastrointestinal and respiratory tracts. Therefore, here, the authors will provide a comprehensive review of the content and functions of different nutritional and bioactive anti-infectious components from breast milk, such as oligosaccharides, lactoferrin, haptocorrin, α-lactalbumin, k-casein, lysozyme, lactoperoxidase, mucin, fatty acids, defensins, cytokines and chemokines, hormones and growth factors, complement proteins, leukocytes and nucleic acids, including microRNAs, among many others, and the induction of antibody responses in breast milk after maternal vaccination with several licensed vaccines, including the anti-SARS-CoV-2 vaccine preparations used worldwide. Currently, in the midst of the pandemic, maternal vaccination has re-emerged as a crucial source of passive immunity to the neonate through the placenta and breastfeeding, considering that maternal vaccination can induce specific antibodies if performed during pregnancy and after delivery. There have been some reports in the literature about milk IgA antibodies induced by bacterial antigens or inactivated virus vaccines, such as anti-diphtheria-tetanus-pertussis, anti-influenza viruses, anti-pneumococcal and meningococcal polysaccharide preparations. Regarding anti-SARS-CoV-2 vaccines, most studies demonstrate elevated levels of specific IgA and IgG antibodies in milk with virus-neutralizing ability after maternal vaccination, which represents an additional approach to improve the protection of the nursling during the entire breastfeeding period.
Collapse
Affiliation(s)
- Yingying Zheng
- Department of Pediatrics, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Simone Correa-Silva
- Department of Pediatrics, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil; Universidade Paulista, UNIP, São Paulo, SP, Brazil.
| | - Patricia Palmeira
- Laboratory of Medical Investigation (LIM-36), Department of Pediatrics, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Magda Carneiro-Sampaio
- Department of Pediatrics, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
32
|
Yang X, Fox A, DeCarlo C, Norris C, Griffin S, Wedekind S, Flanagan JM, Shenker N, Powell RL. Comparative Profiles of SARS-CoV-2 Spike-Specific Human Milk Antibodies Elicited by mRNA- and Adenovirus-Based COVID-19 Vaccines. Breastfeed Med 2022; 17:638-646. [PMID: 35675683 DOI: 10.1089/bfm.2022.0019] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background: Numerous COVID-19 vaccines are authorized globally. To date, ∼71% of doses comprise the Pfizer/BioNTech vaccine, and ∼17% the Moderna/NIH vaccine, both of which are messenger RNA (mRNA) based. The chimpanzee Ad-based Oxford/AstraZeneca (AZ) vaccine comprises ∼9%, while the Johnson & Johnson/Janssen (J&J) human adenovirus (Ad26) vaccine ranks fourth at ∼2%. No COVID-19 vaccine is yet available for children 0-4. One method to protect this population may be passive immunization through antibodies (Abs) provided in the milk of a lactating vaccinated person. Our early work and other reports have demonstrated that unlike the post-SARS-CoV-2 infection milk Ab profile, which is rich in specific secretory (s)IgA, the vaccine response is highly IgG dominant. Results: In this report, we present a comparative assessment of the milk Ab response elicited by Pfizer, Moderna, J&J, and AZ vaccines. This analysis revealed 86-100% of mRNA vaccine recipient milk exhibited Spike-specific IgG endpoint titers, which were 12- to 28-fold higher than those measured for Ad vaccine recipient milk. Ad-based vaccines elicited Spike-specific milk IgG in only 33-38% of recipients. Specific IgA was measured in 52-71% of mRNA vaccine recipient milk and 17-23% of Ad vaccine recipient milk. J&J recipient milk exhibited significantly lower IgA than Moderna recipients, and AZ recipients exhibited significantly lower IgA titers than Moderna and Pfizer. Less than 50% of milk of any group exhibited specific secretory Ab, with Moderna recipient IgA titers measuring significantly higher than AZ. Moderna appeared to most frequently elicit greater than twofold increases in specific secretory Ab titer relative to prevaccine sample. Conclusion: These data indicate that current Ad-based COVID-19 vaccines poorly elicit Spike-specific Ab in milk compared to mRNA-based vaccines, and that mRNA vaccines are preferred for immunizing the lactating population. This study highlights the need to design vaccines better aimed at eliciting an optimal milk Ab response.
Collapse
Affiliation(s)
- Xiaoqi Yang
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alisa Fox
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Claire DeCarlo
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Caroline Norris
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Samantha Griffin
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Sophie Wedekind
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - James M Flanagan
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Natalie Shenker
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Rebecca L Powell
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
33
|
Guo J, Tan M, Zhu J, Tian Y, Liu H, Luo F, Wang J, Huang Y, Zhang Y, Yang Y, Wang G. Proteomic Analysis of Human Milk Reveals Nutritional and Immune Benefits in the Colostrum from Mothers with COVID-19. Nutrients 2022; 14:nu14122513. [PMID: 35745243 PMCID: PMC9227629 DOI: 10.3390/nu14122513] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 02/05/2023] Open
Abstract
Despite the well-known benefits of breastfeeding and the World Health Organization’s breastfeeding recommendations for COVID-19 infected mothers, whether these mothers should be encouraged to breastfeed is under debate due to concern about the risk of virus transmission and lack of evidence of breastmilk’s protective effects against the virus. Here, we provide a molecular basis for the breastfeeding recommendation through mass spectrometry (MS)-based proteomics and glycosylation analysis of immune-related proteins in both colostrum and mature breastmilk collected from COVID-19 patients and healthy donors. The total protein amounts in the COVID-19 colostrum group were significantly higher than in the control group. While casein proteins in COVID-19 colostrum exhibited significantly lower abundances, immune-related proteins, especially whey proteins with antiviral properties against SARS-CoV-2, were upregulated. These proteins were detected with unique site-specific glycan structures and improved glycosylation diversity that are beneficial for recognizing epitopes and blocking viral entry. Such adaptive differences in milk from COVID-19 mothers tended to fade in mature milk from the same mothers one month postpartum. These results suggest that feeding infants colostrum from COVID-19 mothers confers both nutritional and immune benefits, and provide molecular-level insights that aid breastmilk feeding decisions in cases of active infection.
Collapse
Affiliation(s)
- Juanjuan Guo
- Department of Gynaecology and Obstetrics, Zhongnan Hospital of Wuhan University, Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan 430071, China; (J.G.); (H.L.); (Y.Z.)
| | - Minjie Tan
- Institute for Cell Analysis, Shenzhen Bay Laboratory, Shenzhen 518132, China; (M.T.); (Y.T.); (Y.H.)
| | - Jing Zhu
- Institute of Biotechnology and Health, Beijing Academy of Science and Technology, Beijing 100089, China
- Correspondence: (J.Z.); (G.W.)
| | - Ye Tian
- Institute for Cell Analysis, Shenzhen Bay Laboratory, Shenzhen 518132, China; (M.T.); (Y.T.); (Y.H.)
| | - Huanyu Liu
- Department of Gynaecology and Obstetrics, Zhongnan Hospital of Wuhan University, Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan 430071, China; (J.G.); (H.L.); (Y.Z.)
| | - Fan Luo
- State Key Laboratory of Virology, Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China;
| | - Jianbin Wang
- School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China;
| | - Yanyi Huang
- Institute for Cell Analysis, Shenzhen Bay Laboratory, Shenzhen 518132, China; (M.T.); (Y.T.); (Y.H.)
- Biomedical Pioneering Innovation Center, Peking University, Beijing 100871, China
| | - Yuanzhen Zhang
- Department of Gynaecology and Obstetrics, Zhongnan Hospital of Wuhan University, Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan 430071, China; (J.G.); (H.L.); (Y.Z.)
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Yuexin Yang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China;
| | - Guanbo Wang
- Institute for Cell Analysis, Shenzhen Bay Laboratory, Shenzhen 518132, China; (M.T.); (Y.T.); (Y.H.)
- Biomedical Pioneering Innovation Center, Peking University, Beijing 100871, China
- Correspondence: (J.Z.); (G.W.)
| |
Collapse
|
34
|
Demers-Mathieu V, Lavangnananda S. Restricting Cow's Milk in the Maternal Diet Reduces the Titers of β-Lactoglobulin-Specific IgG Antibodies in Human Milk. Breastfeed Med 2022; 17:501-505. [PMID: 35325546 DOI: 10.1089/bfm.2021.0242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Background: Human milk antibodies specific to allergen enhance immunological tolerance in neonates by educating their immature mucosal immunity. The impact of restricting food allergens in diet and maternal factors on the levels of allergen-specific antibodies in human milk remains unclear. We aimed to evaluate the influence of the maternal avoidance diet of cow's milk on the titers of IgG, SIgA/IgA, and IgM specific to β-lactoglobulin (BLG) in human milk. Materials and Methods: Human milk samples were collected from 26 women nonrestricting cow's milk and 11 women restricting cow's milk. The titers of IgG, SIgA/IgA, and SIgM/IgM specific to BLG were measured using ELISA. Results: BLG-specific IgG titers were 2.9-fold higher in women nonrestricting cow's milk than those restricting cow's milk in their diet (p = 0.026), but BLG-specific SIgA/IgA and SIgM/IgM titers were comparable between these two groups. BLG-specific IgG was positively correlated with BLG-specific SIgA/IgA titers in milk from mothers nonrestricting cow's milk (p = 0.0007) but did not correlate for mothers restricting cow's milk. BLG-specific SIgA/IgA titer decreased with increasing postpartum time in milk from women restricting cow's milk (p = 0.019). Type of delivery, infant gender, maternal age, and probiotic intake did not influence the BLG-specific antibody titers. Conclusions: This study reveals that the secretion of BLG-specific IgG in human milk increases in women nonrestricting cow's milk compared with women restricting cow's milk. The role of breast milk allergen-specific antibodies on the neonatal gut (crosstalk with immune and epithelial cells) remains to be investigated.
Collapse
Affiliation(s)
- Veronique Demers-Mathieu
- Medolac Laboratories, A Public Benefit Corporation, Department of Neonatal Immunology and Microbiology, Boulder City, Nevada, USA
| | - Sirima Lavangnananda
- Medolac Laboratories, A Public Benefit Corporation, Department of Neonatal Immunology and Microbiology, Boulder City, Nevada, USA
| |
Collapse
|
35
|
Selma-Royo M, Calvo-Lerma J, Bäuerl C, Esteban-Torres M, Cabrera-Rubio R, Collado MC. Human milk microbiota: what did we learn in the last 20 years? MICROBIOME RESEARCH REPORTS 2022; 1:19. [PMID: 38046359 PMCID: PMC10688795 DOI: 10.20517/mrr.2022.05] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/06/2022] [Accepted: 04/29/2022] [Indexed: 12/05/2023]
Abstract
Human milk (HM) is the gold standard for infant nutrition during the first months of life. Beyond its nutritional components, its complex bioactive composition includes microorganisms, their metabolites, and oligosaccharides, which also contribute to gut colonization and immune system maturation. There is growing evidence of the beneficial effects of bacteria present in HM. However, current research presents limited data on the presence and functions of other organisms. The potential biological impacts on maternal and infant health outcomes, the factors contributing to milk microbes' variations, and the potential functions in the infant's gut remain unclear. This review provides a global overview of milk microbiota, what the actual knowledge is, and what the gaps and challenges are for the next years.
Collapse
Affiliation(s)
| | | | | | | | | | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia 46980, Spain
| |
Collapse
|
36
|
Conti MG, Terreri S, Terrin G, Natale F, Pietrasanta C, Salvatori G, Brunelli R, Midulla F, Papaevangelou V, Carsetti R, Angelidou A. Severe Acute Respiratory Syndrome Coronavirus 2 Infection Versus Vaccination in Pregnancy: Implications for Maternal and Infant Immunity. Clin Infect Dis 2022; 75:S37-S45. [PMID: 35535796 PMCID: PMC9129222 DOI: 10.1093/cid/ciac359] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/22/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been associated with adverse maternal and neonatal outcomes, yet uptake of SARS-CoV-2 vaccines during pregnancy and lactation has been slow. As a result, millions of pregnant and lactating women and their infants remain susceptible to the virus. METHODS We measured spike-specific immunoglobulin G (anti-S IgG) and immunoglobulin A (anti-S IgA) in serum and breastmilk (BM) samples from 3 prospective mother-infant cohorts recruited in 2 academic medical centers. The primary aim was to determine the impact of maternal SARS-CoV-2 immunization vs infection and their timing on systemic and mucosal immunity. RESULTS The study included 28 mothers infected with SARS-CoV-2 in late pregnancy (INF), 11 uninfected mothers who received 2 doses of the BNT162b2 vaccine in the latter half of pregnancy (VAX-P), and 12 uninfected mothers who received 2 doses of BNT162b2 during lactation. VAX dyads had significantly higher serum anti-S IgG compared to INF dyads (P < .0001), whereas INF mothers had higher BM:serum anti-S IgA ratios compared to VAX mothers (P = .0001). Median IgG placental transfer ratios were significantly higher in VAX-P compared to INF mothers (P < .0001). There was a significant positive correlation between maternal and neonatal serum anti-S IgG after vaccination (r = 0.68, P = .013), but not infection. CONCLUSIONS BNT161b2 vaccination in late pregnancy or lactation enhances systemic immunity through serum anti-S immunoglobulin, while SARS-CoV-2 infection induces mucosal over systemic immunity more efficiently through BM immunoglobulin production. Next-generation vaccines boosting mucosal immunity could provide additional protection to the mother-infant dyad. Future studies should focus on identifying the optimal timing of primary and/or booster maternal vaccination for maximal benefit.
Collapse
Affiliation(s)
- Maria Giulia Conti
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Sara Terreri
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Gianluca Terrin
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Fabio Natale
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Carlo Pietrasanta
- NICU, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Guglielmo Salvatori
- Neonatal Intensive Care Unit and Human Milk Bank, Department of Neonatology, Bambino Gesù Children’s Hospital, IRCSS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy
| | - Roberto Brunelli
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Fabio Midulla
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Vassiliki Papaevangelou
- Third Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| | - Rita Carsetti
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy,Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children's Hospital, IRCCS; Piazza Sant’Onofrio, 4, 00165, Rome, Italy
| | - Asimenia Angelidou
- Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, United States,Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital,Harvard Medical School,Corresponding author: Asimenia Angelidou, MD PhD Instructor in Pediatrics, Harvard Medical School 4 Blackfan Circle, HIM Building, Rm 836, Boston MA 02115
| |
Collapse
|
37
|
Pannaraj PS, da Costa-Martins AG, Cerini C, Li F, Wong SS, Singh Y, Urbanski AH, Gonzalez-Dias P, Yang J, Webby RJ, Nakaya HI, Aldrovandi GM. Molecular alterations in human milk in simulated maternal nasal mucosal infection with live attenuated influenza vaccination. Mucosal Immunol 2022; 15:1040-1047. [PMID: 35739193 PMCID: PMC9225800 DOI: 10.1038/s41385-022-00537-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/24/2022] [Accepted: 05/31/2022] [Indexed: 02/04/2023]
Abstract
Breastfeeding protects against mucosal infections in infants. The underlying mechanisms through which immunity develops in human milk following maternal infection with mucosal pathogens are not well understood. We simulated nasal mucosal influenza infection through live attenuated influenza vaccination (LAIV) and compared immune responses in milk to inactivated influenza vaccination (IIV). Transcriptomic analysis was performed on RNA extracted from human milk cells to evaluate differentially expressed genes and pathways on days 1 and 7 post-vaccination. Both LAIV and IIV vaccines induced influenza-specific IgA that persisted for at least 6 months. Regulation of type I interferon production, toll-like receptor, and pattern recognition receptor signaling pathways were highly upregulated in milk on day 1 following LAIV but not IIV at any time point. Upregulation of innate immunity in human milk may provide timely protection against mucosal infections until antigen-specific immunity develops in the human milk-fed infant.
Collapse
Affiliation(s)
- Pia S Pannaraj
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, CA, USA.
- Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA, USA.
| | - André Guilherme da Costa-Martins
- School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
- Scientific Platform Pasteur-University of São Paulo, São Paulo, Brazil
| | - Chiara Cerini
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Fan Li
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Sook-San Wong
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
- School of Public Health, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Youvika Singh
- School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
- Scientific Platform Pasteur-University of São Paulo, São Paulo, Brazil
| | - Alysson H Urbanski
- School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Patrícia Gonzalez-Dias
- Hospital Israelita Albert Einstein, São Paulo, Brazil
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Juliana Yang
- School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Richard J Webby
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Helder I Nakaya
- Scientific Platform Pasteur-University of São Paulo, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Grace M Aldrovandi
- Department of Pediatrics, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
38
|
Richards A, Baranova D, Mantis NJ. The prospect of orally administered monoclonal secretory IgA (SIgA) antibodies to prevent enteric bacterial infections. Hum Vaccin Immunother 2022; 18:1964317. [PMID: 34491878 PMCID: PMC9103515 DOI: 10.1080/21645515.2021.1964317] [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] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/08/2021] [Accepted: 07/30/2021] [Indexed: 12/20/2022] Open
Abstract
Eliminating diarrheal diseases as a leading cause of childhood morbidity and mortality in low- and middle-income countries (LMICs) will require multiple intervention strategies. In this review, we spotlight a series of preclinical studies investigating the potential of orally administered monoclonal secretory IgA (SIgA) antibodies (MAbs) to reduce disease associated with three enteric bacterial pathogens: Campylobacter jejuni, enterotoxigenic Escherichia coli (ETEC), and invasive Salmonella enterica serovar Typhimurium. IgA MAbs targeting bacterial surface antigens (flagella, adhesins, and lipopolysaccharide) were generated from mice, humanized mice, and human tonsillar B cells. Recombinant SIgA1 and/or SIgA2 derivates of those MAbs were purified from supernatants following transient transfection of 293 cells with plasmids encoding antibody heavy and light chains, J-chain, and secretory component (SC). When administered to mice by gavage immediately prior to (or admixed with) the bacterial challenge, SIgA MAbs reduced infection C. jejuni, ETEC, and S. Typhimurium infections. Fv-matched IgG1 MAbs by comparison were largely ineffective against C. jejuni and S. Typhimurium under the same conditions, although they were partially effective against ETEC. While these findings highlight future applications of orally administered SIgA, the studies also underscored the fundamental challenges associated with using MAbs as prophylactic tools against enteric bacterial diseases.
Collapse
Affiliation(s)
- Angelene Richards
- Department of Biomedical Sciences, University at Albany School, Albany, NY, USA
| | - Danielle Baranova
- Department of Biomedical Sciences, University at Albany School, Albany, NY, USA
| | - Nicholas J. Mantis
- Department of Biomedical Sciences, University at Albany School, Albany, NY, USA
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| |
Collapse
|
39
|
Lokossou GAG, Kouakanou L, Schumacher A, Zenclussen AC. Human Breast Milk: From Food to Active Immune Response With Disease Protection in Infants and Mothers. Front Immunol 2022; 13:849012. [PMID: 35450064 PMCID: PMC9016618 DOI: 10.3389/fimmu.2022.849012] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/07/2022] [Indexed: 12/29/2022] Open
Abstract
Breastfeeding is associated with long-term wellbeing including low risks of infectious diseases and non-communicable diseases such as asthma, cancer, autoimmune diseases and obesity during childhood. In recent years, important advances have been made in understanding the human breast milk (HBM) composition. Breast milk components such as, non-immune and immune cells and bioactive molecules, namely, cytokines/chemokines, lipids, hormones, and enzymes reportedly play many roles in breastfed newborns and in mothers, by diseases protection and shaping the immune system of the newborn. Bioactive components in HBM are also involved in tolerance and appropriate inflammatory response of breastfed infants if necessary. This review summarizes the current literature on the relationship between mother and her infant through breast milk with regard to disease protection. We will shed some light on the mechanisms underlying the roles of breast milk components in the maintenance of health of both child and mother.
Collapse
Affiliation(s)
- Gatien A. G. Lokossou
- Research Unit in Applied Microbiology and Pharmacology of Natural Substances, Polytechnic School of Abomey-Calavi, Department Human Biology Engineering, University of Abomey-Calavi, Abomey-Calavi, Benin
| | - Léonce Kouakanou
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Anne Schumacher
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research and Perinatal Immunology, Saxonian Incubator for Clinical Translation, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Ana C. Zenclussen
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research and Perinatal Immunology, Saxonian Incubator for Clinical Translation, Medical Faculty, University of Leipzig, Leipzig, Germany
| |
Collapse
|
40
|
Ahmad HI, Jabbar A, Mushtaq N, Javed Z, Hayyat MU, Bashir J, Naseeb I, Abideen ZU, Ahmad N, Chen J. Immune Tolerance vs. Immune Resistance: The Interaction Between Host and Pathogens in Infectious Diseases. Front Vet Sci 2022; 9:827407. [PMID: 35425833 PMCID: PMC9001959 DOI: 10.3389/fvets.2022.827407] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/17/2022] [Indexed: 11/13/2022] Open
Abstract
The immune system is most likely developed to reduce the harmful impact of infections on the host homeostasis. This defense approach is based on the coordinated activity of innate and adaptive immune system components, which detect and target infections for containment, killing, or expulsion by the body's defense mechanisms. These immunological processes are responsible for decreasing the pathogen burden of an infected host to maintain homeostasis that is considered to be infection resistance. Immune-driven resistance to infection is connected with a second, and probably more important, defensive mechanism: it helps to minimize the amount of dysfunction imposed on host parenchymal tissues during infection without having a direct adverse effect on pathogens. Disease tolerance is a defensive approach that relies on tissue damage control systems to prevent infections from causing harm to the host. It also uncouples immune-driven resistance mechanisms from immunopathology and disease, allowing the body to fight infection more effectively. This review discussed the cellular and molecular processes that build disease tolerance to infection and the implications of innate immunity on those systems. In addition, we discuss how symbiotic relationships with microbes and their control by particular components of innate and adaptive immunity alter disease tolerance to infection.
Collapse
Affiliation(s)
- Hafiz Ishfaq Ahmad
- Department of Animal Breeding and Genetics, University of Veterinary and Animal Sciences, Lahore, Pakistan
- *Correspondence: Hafiz Ishfaq Ahmad
| | - Abdul Jabbar
- Department of Clinical Medicine, Faculty of Veterinary Science, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Nadia Mushtaq
- Department of Biological Sciences, Faculty of Fisheries and Wildlife, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Zainab Javed
- Institute of Pharmaceutical Sciences, Faculty of Biosciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Muhammad Umar Hayyat
- Institute of Pharmaceutical Sciences, Faculty of Biosciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Javaria Bashir
- Department of Medical Sciences, Sharif Medical and Dental Hospital, Lahore, Pakistan
| | - Iqra Naseeb
- Institute of Microbiology, Faculty of Veterinary Science, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Zain Ul Abideen
- Department of Zoology, Ghazi University, Dera Ghazi Khan, Pakistan
| | - Nisar Ahmad
- Department of Livestock Management, University of Veterinary and Animal Sciences, Pattoki, Pakistan
| | - Jinping Chen
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China
- Jinping Chen
| |
Collapse
|
41
|
Fox A, Marino J, Amanat F, Oguntuyo KY, Hahn-Holbrook J, Lee B, Zolla-Pazner S, Powell RL. The IgA in milk induced by SARS-CoV-2 infection is comprised of mainly secretory antibody that is neutralizing and highly durable over time. PLoS One 2022; 17:e0249723. [PMID: 35263323 PMCID: PMC8906612 DOI: 10.1371/journal.pone.0249723] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 01/28/2022] [Indexed: 12/12/2022] Open
Abstract
Approximately 10% of infants infected with SARS-CoV-2 will experience COVID-19 illness requiring advanced care. A potential mechanism to protect this population is passive immunization via the milk of a previously infected person. We and others have reported on the presence of SARS-CoV-2-specific antibodies in human milk. We now report the prevalence of SARS-CoV-2 IgA in the milk of 74 COVID-19-recovered participants, and find that 89% of samples are positive for Spike-specific IgA. In a subset of these samples, 95% exhibited robust IgA activity as determined by endpoint binding titer, with 50% considered high-titer. These IgA-positive samples were also positive for Spike-specific secretory antibody. Levels of IgA antibodies and secretory antibodies were shown to be strongly positively correlated. The secretory IgA response was dominant among the milk samples tested compared to the IgG response, which was present in 75% of samples and found to be of high-titer in only 13% of cases. Our IgA durability analysis using 28 paired samples, obtained 4–6 weeks and 4–10 months after infection, found that all samples exhibited persistently significant Spike-specific IgA, with 43% of donors exhibiting increasing IgA titers over time. Finally, COVID-19 and pre-pandemic control milk samples were tested for the presence of neutralizing antibodies; 6 of 8 COVID-19 samples exhibited neutralization of Spike-pseudotyped VSV (IC50 range, 2.39–89.4ug/mL) compared to 1 of 8 controls. IgA binding and neutralization capacities were found to be strongly positively correlated. These data are highly relevant to public health, not only in terms of the protective capacity of these antibodies for breastfed infants, but also for the potential use of such antibodies as a COVID-19 therapeutic, given that secretory IgA is highly in all mucosal compartments.
Collapse
Affiliation(s)
- Alisa Fox
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Jessica Marino
- Health Sciences Research Institute, University of California Merced, Merced, California, United States of America
- Department of Psychology, University of California Merced, Merced, California, United States of America
| | - Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Kasopefoluwa Y. Oguntuyo
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Jennifer Hahn-Holbrook
- Health Sciences Research Institute, University of California Merced, Merced, California, United States of America
- Department of Psychology, University of California Merced, Merced, California, United States of America
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Susan Zolla-Pazner
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Rebecca L. Powell
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
42
|
Winingsih GAM, Salmah U, Masni M, Indriasari R, Karmaya INM, Amiruddin R, Nasir S, Birawida AB. Effect of Co-parenting on the Prevention of Postpartum Blues and Quality of Breast Milk: A Literature Review. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.8385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
INTRODUCTION: Breast milk is the best food and nutrition for a baby’s development and also provides natural immunity; hence, the World Health Organization and the Indonesian Ministry of Health recommend exclusive breastfeeding for up to 6 months. The quality and duration of breastfeeding are very important, but there are several influencing factors such as maternal self-efficacy, fatigue, techniques, and life partner. Meanwhile, co-parenting is the right method in maintaining the continuity and quality of breastfeeding.
AIM: This study aims to explore literature related to co-parenting and its relation to postpartum blues in improving the quality of breast milk.
METHODS: A literature search was carried out using three databases, namely, PubMed (n = 28), ScienceDirect (n = 22), ProQuest (n = 108) and the results were selected based on the inclusion and exclusion criteria in line with PRISMA guidelines in five articles.
RESULTS: The screening obtained five articles written in the past 10 years, English, full text, and with open access for review. Of the five articles that we reviewed, all of them support co-parenting in preventing postpartum and quality of breast milk, several studies that we reviewed found that co-parenting supports the improvement and process of breastfeeding in mothers.
CONCLUSION: Based on the results, co-parenting is the right method to improve the quality and sustainability of breast milk to prevent the occurrence of postpartum blues; hence, it is recommended to be implemented.
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW One important question from the outset of the pandemic has been whether a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected person's milk might be a vehicle for SARS-CoV-2 transmission. This review summarizes the most recent data on this topic. RECENT FINDINGS A SARS-CoV-2 sIgA response in milk after infection is very common. To date, there has been no evidence that SARS-CoV-2 transmits via human milk. Though viral RNA has been identified in a minority of milk samples studied, infectious virus particles have not. SUMMARY The highly dominant transmission route for SARS-CoV-2 is via inhalation of respiratory droplets containing virus particles. Other routes of transmission are possible, including fecal-oral, trans-placental, and to a much lesser extent, via a contaminated surface. SARS-CoV-2 cannot transmit via human milk. There is no evidence that infants should be separated from SARS-CoV-2-infected mothers who are well enough to establish or continue breastfeeding.
Collapse
Affiliation(s)
- Rebecca L R Powell
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
44
|
Chen LL, Liu J, Mu XH, Zhang XY, Yang CZ, Xiong XY, Wang MQ. Oropharyngeal administration of mother's own milk influences levels of salivary sIgA in preterm infants fed by gastric tube. Sci Rep 2022; 12:2233. [PMID: 35140309 PMCID: PMC8828761 DOI: 10.1038/s41598-022-06243-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/25/2022] [Indexed: 11/21/2022] Open
Abstract
The aim of the present study was to explore the effect of oropharyngeal mother’s milk administration on salivary secretory immunoglobulin A (sIgA) levels in preterm infants fed by gastric tube. Infants (n = 130) with birth weight < 1500 g were randomly allocated into two groups which both received breast milk for enteral nutrition. The experimental group (n = 65) accepted oropharyngeal mother’s milk administration before gastric tube feeding for 14 days after birth. The control group (n = 65) accepted oropharyngeal 0.9% normal saline administration. Saliva concentration of sIgA were assessed at the 2 h, 7th and 14th day after birth. The level of salivary sIgA in experimental group were significantly higher than those in control group on the 7th day after birth (p < 0.05), but there were no differences in salivary sIgA levels on the 14th day between the two groups. The results of quantile regression analysis showed that oropharyngeal mother’s milk administration, delivery mode and gestational age had significant effects on the increase of sIgA. SIgA in experimental group and the total number of intervention had a significant positive correlation (p < 0.05). Oropharyngeal mother’s milk administration can improve salivary sIgA levels of preterm infants.
Collapse
Affiliation(s)
- Li-Lian Chen
- Department of Neonatology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Jie Liu
- Shanxi University of Chinese Medicine, Shanxi, China
| | - Xiao-He Mu
- Shanxi University of Chinese Medicine, Shanxi, China
| | - Xi-Yang Zhang
- Shanxi University of Chinese Medicine, Shanxi, China
| | - Chuan-Zhong Yang
- Department of Neonatology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China.
| | - Xiao-Yun Xiong
- Department of Neonatology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Mei-Qi Wang
- Department of Neonatology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| |
Collapse
|
45
|
Singh G, Tucker EW, Rohlwink UK. Infection in the Developing Brain: The Role of Unique Systemic Immune Vulnerabilities. Front Neurol 2022; 12:805643. [PMID: 35140675 PMCID: PMC8818751 DOI: 10.3389/fneur.2021.805643] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 12/30/2021] [Indexed: 11/13/2022] Open
Abstract
Central nervous system (CNS) infections remain a major burden of pediatric disease associated with significant long-term morbidity due to injury to the developing brain. Children are susceptible to various etiologies of CNS infection partly because of vulnerabilities in their peripheral immune system. Young children are known to have reduced numbers and functionality of innate and adaptive immune cells, poorer production of immune mediators, impaired responses to inflammatory stimuli and depressed antibody activity in comparison to adults. This has implications not only for their response to pathogen invasion, but also for the development of appropriate vaccines and vaccination strategies. Further, pediatric immune characteristics evolve across the span of childhood into adolescence as their broader physiological and hormonal landscape develop. In addition to intrinsic vulnerabilities, children are subject to external factors that impact their susceptibility to infections, including maternal immunity and exposure, and nutrition. In this review we summarize the current evidence for immune characteristics across childhood that render children at risk for CNS infection and introduce the link with the CNS through the modulatory role that the brain has on the immune response. This manuscript lays the foundation from which we explore the specifics of infection and inflammation within the CNS and the consequences to the maturing brain in part two of this review series.
Collapse
Affiliation(s)
- Gabriela Singh
- Division of Neurosurgery, Department of Surgery, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Elizabeth W. Tucker
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ursula K. Rohlwink
- Division of Neurosurgery, Department of Surgery, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Francis Crick Institute, London, United Kingdom
| |
Collapse
|
46
|
Cai M, Lin L, Peng Y, Chen L, Lin Y. Effect of Breast Milk Oral Care on Mechanically Ventilated Preterm Infants: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Front Pediatr 2022; 10:899193. [PMID: 35874566 PMCID: PMC9301042 DOI: 10.3389/fped.2022.899193] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/07/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The benefits of breast milk oral care in mechanically ventilated preterm infants remain controversial. This study aimed to systematically review the evidence on the benefits of breast milk oral care in mechanically ventilated preterm infants. METHODS The randomized controlled trials of breast milk oral care for mechanically ventilated preterm infants were searched in EMBASE, PubMed, Cochrane Library, Web of Science, WANFANG Date and China National Knowledge Infrastructure databases. The retrieval language was limited to Chinese and English, and the final search was conducted until March 2022. Outcome measures included ventilator-associated pneumonia (VAP), mechanical ventilation time (MVT), length of stay (LOS), necrotizing enterocolitis (NEC), late-onset sepsis, mortality during hospitalization, time of full intestinal feeding and time of full oral feeding. Two researchers independently screened the literature, extracted the data, and conducted the literature quality assessment. Meta-analysis was mainly performed using RevMan 5.3. RESULTS Eight articles involving 1,046 preterm infants were included. Our meta-analysis showed that compared with the control group, breast milk oral care could reduce the incidence of VAP [RR = 0.41, 95% CI (0.23, 0.75), P = 0.003] and NEC [RR = 0.54, 95% CI (0.30, 0.95), P = 0.03], and shorten the MVT [MD = -0.45, 95% CI (-0.73, -0.18), P = 0.001] and LOS [MD = -5.74, 95% CI (-10.39, -1.10), P = 0.02]. There were no significant differences in the mortality during hospitalization [RR = 0.94, 95% CI (0.67, 1.33), P = 0.74], the incidence of late-onset sepsis [RR = 0.79, 95% CI (0.40, 1.59), P = 0.51], the time of full intestinal feeding [MD = -2.42, 95% CI (-5.37, 0.52), P = 0.11] and the time of full oral feeding [MD = -3.40, 95% CI (-10.70, 3.91), P = 0.36] between the two groups. CONCLUSIONS Oral care of breast milk can reduce the incidence of VAP and NEC, shorten MVT and LOS in mechanically ventilated preterm infants. However, due to the quality and quantity limitations of the included studies, larger sample size and more strictly designed clinical trials are still needed in the future to further confirm the findings of this study.
Collapse
Affiliation(s)
- Meiling Cai
- Department of Cardiac Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Lingyu Lin
- Department of Nursing, Union Hospital, Fujian Medical University, Fuzhou, China.,School of Nursing, Fujian Medical University, Fuzhou, China
| | - Yanchun Peng
- Department of Cardiac Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Liangwan Chen
- Department of Cardiac Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Yanjuan Lin
- Department of Cardiac Surgery, Union Hospital, Fujian Medical University, Fuzhou, China.,Department of Nursing, Union Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
47
|
Kato I, Horike K, Kawada K, Htun Y, Nishida T, Nakamura S, Koyano K, Konishi Y, Kusaka T. The Trajectory of Expressed Colostrum Volume in the First 48 Hours Postpartum: An Observational Study. Breastfeed Med 2022; 17:52-58. [PMID: 34529518 DOI: 10.1089/bfm.2020.0366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Objective: Colostrum, the first form of human milk, is strongly encouraged for infants due to its benefits. During the early postpartum (PP) period, the secreted colostrum volume can be minimal, causing concerns among mothers about sufficient milk supply. Few studies have examined temporal changes in the colostrum. This study aimed to elucidate the trajectory of expressed colostrum volume in the first 48 hours after delivery. Materials and Methods: This was a cross-sectional observational study performed at Kagawa National Children's Hospital. One hundred five mothers who did not directly breastfeed in the first 48 hours after delivery were enrolled in the study. Well-trained midwives instructed the mothers on how to express human milk, and mothers started to express as soon as possible after delivery. Mothers were advised to express human milk every 3 hours, and the milk volume was measured. Results: Within 3 hours PP, 60% of mothers expressed milk, and the median frequency of expression was 14 (interquartile range, 11-16) times in the first 48 hours. At 0-3 and 3-6 hours PP, the volume of initially expressed milk was 0.4 (0.0-2.0) mL and 1.0 (0.0-6.0) mL, respectively. Subsequently, milk volume decreased. The volume remained low until 30 hours PP and increased dramatically; this phenomenon is termed secretory activation, which began later in primiparous women than in multiparous women. Conclusion: The decline in expressed milk volume during the early PP period caused concern among mothers. Therefore, mothers should be informed of the PP trajectory of human milk volume.
Collapse
Affiliation(s)
- Ikuko Kato
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Kimiyo Horike
- Department of Pediatrics, National Kagawa Children's Hospital, Zentsuji, Japan
| | - Kou Kawada
- Department of Pediatrics, National Kagawa Children's Hospital, Zentsuji, Japan.,Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan
| | - Yinmon Htun
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Tomoko Nishida
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Shinji Nakamura
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Kosuke Koyano
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Yukihiko Konishi
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Takashi Kusaka
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| |
Collapse
|
48
|
Juncker HG, Mulleners SJ, van Gils MJ, Bijl TPL, de Groot CJM, Pajkrt D, Korosi A, van Goudoever JB, van Keulen BJ. Comparison of SARS-CoV-2-Specific Antibodies in Human Milk after mRNA-Based COVID-19 Vaccination and Infection. Vaccines (Basel) 2021; 9:vaccines9121475. [PMID: 34960222 PMCID: PMC8706455 DOI: 10.3390/vaccines9121475] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/03/2021] [Accepted: 12/08/2021] [Indexed: 12/30/2022] Open
Abstract
SARS-CoV-2-specific antibodies are secreted into human milk of infected or vaccinated lactating women and might provide protection to the breastfed infant against COVID-19. Differences in antibody response after these types of exposure are unknown. In this longitudinal cohort study, we compared the antibody response in human milk following SARS-CoV-2 vaccination or infection. We analyzed 448 human milk samples of 28 lactating women vaccinated with the SARS-CoV-2 vaccine BNT162b2 as well as 82 human milk samples of 18 lactating women with a prior SARS-CoV-2 infection. The levels of SARS-CoV-2-specific IgA in human milk were determined over a period of 70 days both after vaccination and infection. The amount of SARS-CoV-2-specific IgA in human milk was similar after SARS-CoV-2 vaccination and infection. After infection, the variability in IgA levels was higher than after vaccination. Two participants with detectable IgA prior to vaccination were analyzed separately and showed higher IgA levels following vaccination compared to both groups. In conclusion, breastfed infants of mothers who have been vaccinated with the BNT162b2 vaccine receive human milk with similar amounts of SARS-CoV-2-specific antibodies compared to infants of previously infected mothers.
Collapse
Affiliation(s)
- Hannah G. Juncker
- Department of Pediatrics, Emma Children’s Hospital, Amsterdam Reproduction & Development Research Institute, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands; (H.G.J.); (S.J.M.); (D.P.); (B.J.v.K.)
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands;
| | - Sien J. Mulleners
- Department of Pediatrics, Emma Children’s Hospital, Amsterdam Reproduction & Development Research Institute, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands; (H.G.J.); (S.J.M.); (D.P.); (B.J.v.K.)
| | - Marit J. van Gils
- Department of Medical Microbiology and Infection Prevention, Amsterdam Infection and Immunity Institute, University of Amsterdam, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands; (M.J.v.G.); (T.P.L.B.)
| | - Tom P. L. Bijl
- Department of Medical Microbiology and Infection Prevention, Amsterdam Infection and Immunity Institute, University of Amsterdam, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands; (M.J.v.G.); (T.P.L.B.)
| | - Christianne J. M. de Groot
- Department of Obstetrics and Gynaecology, Amsterdam Reproduction & Development Research Institute, Vrije Universiteit, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands;
| | - Dasja Pajkrt
- Department of Pediatrics, Emma Children’s Hospital, Amsterdam Reproduction & Development Research Institute, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands; (H.G.J.); (S.J.M.); (D.P.); (B.J.v.K.)
| | - Aniko Korosi
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands;
| | - Johannes B. van Goudoever
- Department of Pediatrics, Emma Children’s Hospital, Amsterdam Reproduction & Development Research Institute, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands; (H.G.J.); (S.J.M.); (D.P.); (B.J.v.K.)
- Correspondence: ; Tel.: +31-20-566-8885
| | - Britt J. van Keulen
- Department of Pediatrics, Emma Children’s Hospital, Amsterdam Reproduction & Development Research Institute, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands; (H.G.J.); (S.J.M.); (D.P.); (B.J.v.K.)
| |
Collapse
|
49
|
Tsugami Y, Ishiba Y, Suzuki N, Nii T, Kobayashi K, Isobe N. Local Heat Treatment of Goat Udders Influences Innate Immune Functions in Mammary Glands. J Mammary Gland Biol Neoplasia 2021; 26:387-397. [PMID: 35015201 DOI: 10.1007/s10911-022-09509-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
Heat stress and mastitis adversely affect milk production in dairy ruminants. Although the udder temperature is elevated in both conditions, the influence of this local temperature rise on milk production and immune function of ruminant mammary glands remains unclear. To address this question, we heated the mammary glands of goats by covering one half of the udder with a disposable heating pad for 24 h, the other uncovered half served as a control. Sixteen Tokara goats (1-5 parity) and three Shiba goats (1-2 parity) at the mid-lactation stage were individually housed, fed 0.6 kg of hay cubes and 0.2 kg of barley per day, and had free access to water and trace-mineralized salt blocks. Milk samples were collected every 6 h for 24 h after covering (n = 16), and deep mammary gland tissue areas were collected after 24 h (n = 5). The concentrations of antimicrobial components [lactoferrin, β-defensin-1, cathelicidin-2, cathelicidin-7, and immunoglobulin A (IgA)] in milk were measured by the enzyme-linked immunosorbent assay (ELISA). The localization of IgA was examined by immunohistochemistry. The mRNA expression and protein concentrations of C-C motif chemokine ligand-28 (CCL-28) and interleukin (IL)-8 in the mammary gland tissue were measured using quantitative polymerase chain reaction and ELISA, respectively. The somatic cell count in milk was significantly higher in the local heat-treatment group than in the control group after 12 h of treatment. The treatment group had significantly higher concentrations of cathelicidin-2 and IgA than the control group after 24 h of treatment. In addition, the number of IgA-positive cells in the mammary stromal region and the concentration of CCL-28 in the mammary glands were increased by local heat treatment. In conclusion, a local rise in udder temperature enhanced the innate immune function in mammary glands by increasing antimicrobial components.
Collapse
Affiliation(s)
- Yusaku Tsugami
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama Higashi-Hiroshima, Hiroshima, 739-8528, Japan
| | - Yuki Ishiba
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama Higashi-Hiroshima, Hiroshima, 739-8528, Japan
| | - Naoki Suzuki
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama Higashi-Hiroshima, Hiroshima, 739-8528, Japan
| | - Takahiro Nii
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama Higashi-Hiroshima, Hiroshima, 739-8528, Japan
| | - Ken Kobayashi
- Research Faculty of Agriculture, Hokkaido University, North 9, West 9, Sapporo, 060-8589, Japan
| | - Naoki Isobe
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama Higashi-Hiroshima, Hiroshima, 739-8528, Japan.
| |
Collapse
|
50
|
Effect of breast milk oral care in infants who underwent surgical correction of ventricular septal defect. Cardiol Young 2021; 31:2015-2018. [PMID: 33883048 DOI: 10.1017/s1047951121001438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE This study explored the clinical effect of employing breast milk oral care for infants who underwent surgical correction of ventricular septal defect. METHODS A prospective randomised controlled study was conducted in a provincial hospital between January, 2020 and July, 2020 in China. Patients were randomly divided into an intervention group (breast milk oral care, n = 28) and a control group (physiological saline oral care, n = 28). The intervention group was given oral nursing using breast milk for infants in the early post-operative period, and the control group was given oral nursing using physiological saline. Related clinical data were recorded and analysed. RESULTS There were no significant differences in age, gender, weight, operation time, cardiopulmonary bypass time, or aortic cross-clamping time between the two groups. Compared with the physiological saline oral care group, the mechanical ventilation duration, the length of ICU stay in the breast milk oral care group were significantly shorter. The time of start feeding and total enteral nutrition were significantly earlier in the intervention group than those in the control group. The incidence of post-operative pneumonia in the breast milk oral care group was 3.6%, which was significantly lower than that of the physiological saline oral care group. CONCLUSION The use of breast milk for oral care in infants who underwent surgical correction of VSD can reduce the incidence of post-operative pneumonia and promote the recovery of gastrointestinal function.
Collapse
|