1
|
Hamdan TA, Alkhateeb S, Oriquat G, Alzoubi A, Ahmed KAA. Impact of breastfeeding and formula feeding on immune cell populations and blood cell parameters: an observational study. J Int Med Res 2024; 52:3000605241307217. [PMID: 39731441 DOI: 10.1177/03000605241307217] [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: 12/29/2024] Open
Abstract
OBJECTIVE Breastfeeding is associated with improved health outcomes in infancy and throughout adulthood as breast milk encompasses diverse immune-active factors that affect the ontogeny of the immune system in breastfed (BF) infants. Nevertheless, the impact of infant feeding on the immune system is poorly understood, and a comprehensive understanding of immune system development in human infants is lacking. In this observational study, we addressed the effects of different infant feeding approaches on cell populations and parameters in the peripheral blood of infants to gain insight into the innate and adaptive arms of the immune system. METHODS Using flowcytometric analysis, we performed complete blood counts and immunoprofiling of peripheral blood collected from BF and formula-fed (FF) infants at different ages. RESULTS Our results showed that the blood of BF infants had a higher frequency of leukocytes and erythrocytes in early infancy. The hemoglobin concentration was enhanced in BF infants. However, the platelet count was comparable regardless of feeding regimen. CONCLUSIONS We observed immunophenotypic differences between the two populations of infants, mirrored by improved frequency of innate and adaptive immune cells in BF infants.
Collapse
Affiliation(s)
- Thamer A Hamdan
- Department of Basic Dental Sciences, Faculty of Dentistry, Al-Ahliyya Amman University, Amman, Jordan
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, Jordan
| | | | - Ghaleb Oriquat
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, Jordan
| | | | - Khaled Abdul-Aziz Ahmed
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, Jordan
| |
Collapse
|
2
|
Soto Ocaña J, Friedman ES, Keenan O, Bayard NU, Ford E, Tanes C, Munneke MJ, Beavers WN, Skaar EP, Bittinger K, Zemel BS, Wu GD, Zackular JP. Metal availability shapes early life microbial ecology and community succession. mBio 2024; 15:e0153424. [PMID: 39440978 PMCID: PMC11558993 DOI: 10.1128/mbio.01534-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
The gut microbiota plays a critical role in human health and disease. Microbial community assembly and succession early in life are influenced by numerous factors. In turn, assembly of this microbial community is known to influence the host, including immune system development, and has been linked to outcomes later in life. To date, the role of host-mediated nutritional immunity and metal availability in shaping microbial community assembly and succession early in life has not been explored in depth. Using a human infant cohort, we show that the metal-chelating protein calprotectin is highly abundant in infants. Taxa previously shown to be successful early colonizers of the infant gut, such as Enterococcus, Enterobacteriaceae, and Bacteroides, are highly resistant to experimental metal starvation in culture. Lactobacillus, meanwhile, is highly susceptible to metal restriction, pointing to a possible mechanism by which host-mediated metal limitation shapes the fitness of early colonizing taxa in the infant gut. We further demonstrate that formula-fed infants harbor markedly higher levels of metals in their gastrointestinal tract compared to breastfed infants. Formula-fed infants with high levels of metals harbor distinct microbial communities compared to breastfed infants, with higher levels of Enterococcus, Enterobacter, and Klebsiella, taxa which show increased resistance to the toxic effects of high metal concentrations. These data highlight a new paradigm in microbial community assembly and suggest an unappreciated role for nutritional immunity and dietary metals in shaping the earliest colonization events of the microbiota.IMPORTANCEEarly life represents a critical window for microbial colonization of the human gastrointestinal tract. Surprisingly, we still know little about the rules that govern the successful colonization of infants and the factors that shape the success of early life microbial colonizers. In this study, we report that metal availability is an important factor in the assembly and succession of the early life microbiota. We show that the host-derived metal-chelating protein, calprotectin, is highly abundant in infants and successful early life colonizers can overcome metal restriction. We further demonstrate that feeding modality (breastmilk vs formula) markedly impacts metal levels in the gut, potentially influencing microbial community succession. Our work suggests that metals, a previously unexplored aspect of early life ecology, may play a critical role in shaping the early events of microbiota assembly in infants.
Collapse
Affiliation(s)
- Joshua Soto Ocaña
- Division of Protective Immunity, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elliot S. Friedman
- Division of Gastroenterology & Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Orlaith Keenan
- Division of Protective Immunity, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nile U. Bayard
- Division of Protective Immunity, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Eileen Ford
- Division of Gastroenterology, Hepatology and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ceylan Tanes
- Division of Gastroenterology, Hepatology and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Matthew J. Munneke
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - William N. Beavers
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Eric P. Skaar
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Kyle Bittinger
- Division of Gastroenterology, Hepatology and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- The Center for Microbial Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Babette S. Zemel
- Division of Gastroenterology, Hepatology and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Gary D. Wu
- Division of Gastroenterology & Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joseph P. Zackular
- Division of Protective Immunity, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- The Center for Microbial Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
3
|
Luo F, Zhang M, Zhang L, Zhou P. Nutritional and health effects of bovine colostrum in neonates. Nutr Rev 2024; 82:1631-1645. [PMID: 38052234 DOI: 10.1093/nutrit/nuad145] [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: 12/07/2023] Open
Abstract
High concentrations of immunoglobulins, bioactive peptides, and growth factors are found in bovine colostrum (BC), the milk produced by cows in the first few days after parturition. Various biological functions make it increasingly used to provide nutritional support and immune protection to the offspring of many species, including humans. These biological functions include cell growth stimulation, anti-infection, and immunomodulation. The primary components and biological functions of colostrum were reviewed in the literature, and the authors also looked at its latent effects on the growth and development of neonates as well as on conditions such as infections, necrotizing enterocolitis, short bowel syndrome, and feeding intolerance. The importance of BC in neonatal nutrition, immune support, growth and development, and gut health has been demonstrated in a number of experimental and animal studies. BC has also been shown to be safe at low doses without adverse effects in newborns. BC supplementation has been shown to be efficient in preventing several disorders, including rotavirus diarrhea, necrotizing enterocolitis, and sepsis in animal models of prematurity and some newborn studies. Therefore, BC supplementation should be considered in cases where maternal milk is insufficient or donor milk is unavailable. The optimal age, timing, dosage, and form of BC administration still require further investigation.
Collapse
Affiliation(s)
- Fangmei Luo
- Department of Neonatology, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Min Zhang
- Department of Neonatology, Jinan University-Affiliated Shenzhen Baoan Women's and Children's Hospital, Shenzhen, China
| | - Lian Zhang
- Department of Neonatology, Jinan University-Affiliated Shenzhen Baoan Women's and Children's Hospital, Shenzhen, China
| | - Ping Zhou
- Department of Neonatology, Jinan University-Affiliated Shenzhen Baoan Women's and Children's Hospital, Shenzhen, China
| |
Collapse
|
4
|
Tran LC, Marousez L, Micours E, De Lamballerie M, Thys L, Gottrand F, Ley D, Lesage J, Titécat M. High hydrostatic pressure is similar to Holder pasteurization in preserving donor milk antimicrobial activity. Pediatr Res 2024; 95:1749-1753. [PMID: 38280953 DOI: 10.1038/s41390-024-03022-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 12/22/2023] [Accepted: 12/29/2023] [Indexed: 01/29/2024]
Abstract
BACKGROUND The microbiological safety of donor milk (DM) is commonly ensured by Holder pasteurization (HoP, 62.5 °C for 30 min) in human milk banks despite its detrimental effects on bioactive factors. We compared the antimicrobial properties of DM after Holder pasteurization treatment or High Hydrostatic Pressure processing (HHP, 350 MPa at 38 °C), a non-thermal substitute for DM sterilization. METHODS We assessed lactoferrin and lysozyme concentrations in raw, HHP- and HoP-treated pools of DM (n = 8). The impact of both treatments was evaluated on the growth of Escherichia coli and Group B Streptococcus in comparison with control media (n = 4). We also addressed the effect of storage of HHP treated DM over a 6-month period (n = 15). RESULTS HHP milk demonstrated similar concentrations of lactoferrin compared with raw milk, while it was significantly decreased by HoP. Lysozyme concentrations remained stable regardless of the condition. Although a bacteriostatic effect was observed against Escherichia coli at early timepoints, a sharp bactericidal effect was observed against Group B Streptococcus. Unlike HoP, these results were significant for HHP compared to controls. Stored DM was well and safely preserved by HHP. CONCLUSION Our study demonstrates that this alternative sterilization method shows promise for use with DM in human milk banks. IMPACT Antimicrobial activity of donor milk after High Hydrostatic Pressure treatment has not been clearly evaluated. Donor milk lactoferrin is better preserved by High Hydrostatic Pressure than conventional Holder pasteurization, while lysozyme concentration is not affected by either treatment. As with Holder pasteurization, High Hydrostatic Pressure preserves donor milk bacteriostatic activity against E. coli in addition to bactericidal activity against Group B Streptococcus. Donor milk treated by High Hydrostatic Pressure can be stored safely for 6 months.
Collapse
Affiliation(s)
- Léa Chantal Tran
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France.
- Division of Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Jeanne de Flandre Children's Hospital, CHU Lille, F-59000, Lille, France.
| | - Lucie Marousez
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
| | - Edwina Micours
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
| | | | - Lou Thys
- Laboratory of Bacteriology, Institute of Microbiology, CHU Lille, Lille, France
| | - Frédéric Gottrand
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
- Division of Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Jeanne de Flandre Children's Hospital, CHU Lille, F-59000, Lille, France
| | - Delphine Ley
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
- Division of Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Jeanne de Flandre Children's Hospital, CHU Lille, F-59000, Lille, France
| | - Jean Lesage
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
| | - Marie Titécat
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
- Laboratory of Bacteriology, Institute of Microbiology, CHU Lille, Lille, France
| |
Collapse
|
5
|
Liu Y, Yu X, Zhang G, Xie C, Li Y, Mu P, Chen S, Chen Y, Huang S. Preterm Birth and Infantile Appendicitis. Pediatrics 2023; 152:e2023063815. [PMID: 38018230 DOI: 10.1542/peds.2023-063815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/15/2023] [Indexed: 11/30/2023] Open
Abstract
OBJECTIVE To investigate the potential association between preterm birth and infantile appendicitis. METHODS We conducted a retrospective, multicenter, matched case-control study. This study included consecutive patients <1 year of age with surgery- or autopsy-confirmed appendicitis, admitted between December 2007 and May 2023. For each case, 10 healthy infants were randomly selected and matched by age. Infants were categorized as neonates (0 to 28 days) or older infants (>28 days and <1 year). RESULTS The study included 106 infants diagnosed with appendicitis (median age 2.4 months) and 1060 age-matched healthy controls. In the univariate analysis, preterm birth was significantly associated with the development of appendicitis within the first year of life (odds ratio [OR], 4.23; 95% confidence interval [CI], 2.67-6.70). Other factors associated with a higher risk of infantile appendicitis included being male (OR, 1.91; 95%CI, 1.25-2.94), weight-for-age z-score (OR, 0.72; 95%CI, 0.64-0.81), and exclusively fed on formula (OR, 2.95; 95%CI, 1.77-4.91). In multivariable analyses, preterm remained significantly associated with appendicitis (adjusted OR, 3.32; 95%CI, 1.76-6.24). Subgroup analysis revealed that a preterm birth history increased the risk of appendicitis in both neonates (adjusted OR, 4.56; 95%CI, 2.14-9.71) and older infants (adjusted OR, 3.63; 95%CI, 1.72-7.65). However, preterm did not significantly influence the incidence of appendiceal perforation. CONCLUSIONS Preterm infants have an increased risk of appendicitis during the first year of life. A preterm birth history may help improve the timely diagnosis of infantile appendicitis.
Collapse
Affiliation(s)
- Yakun Liu
- General Surgery Department, Children's Hospital of Soochow University, Soochow, Jiangsu, China
| | - Xiaoxiao Yu
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Guoqing Zhang
- Department of Pediatric Surgery, Weifang People's Hospital, Weifang, Shandong, China
| | - Chuanping Xie
- Department of General Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yang Li
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Pengfei Mu
- Department of Neonatal Surgery, Jinan Children's Hospital, Jinan, Shandong, China
- Department of Pediatric Surgery, Children's Hospital of Shandong University, Jinan, Shandong, China
| | - Shuai Chen
- Department of Neonatal Surgery, Jinan Children's Hospital, Jinan, Shandong, China
- Department of Pediatric Surgery, Children's Hospital of Shandong University, Jinan, Shandong, China
| | - Yajun Chen
- Department of General Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Shungen Huang
- General Surgery Department, Children's Hospital of Soochow University, Soochow, Jiangsu, China
| |
Collapse
|
6
|
Salvatori G, De Rose DU, Clemente M, Gentili C, Verardi GP, Amadio P, Reposi MP, Bagolan P, Dotta A. How much does a liter of donor human milk cost? Cost analysis of operating a human milk bank in Italy. Int Breastfeed J 2022; 17:90. [PMID: 36539788 PMCID: PMC9764658 DOI: 10.1186/s13006-022-00530-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND To date, 40 Human Milk Banks (HMB) have been established in Italy; however, recent cost analysis data for operating an HMB in Italy are not available in the literature. METHODS This study was a cross-sectional study performed at "Bambino Gesù" Children's Hospital in Rome, Italy in 2019. We assessed the one-year operational costs and, the per liter unit costs at our HMB. RESULTS During the 2019 year we collected 771 l of human milk supplied by 128 donors. The total cost was € 178,287.00 and the average cost was € 231.00 per liter. € 188,716.00 would have been spent had the maximum capacity for 904 l been reached. We found a significant difference (€ 231.00 vs € 209.00 per liter, p = 0.016) comparing the cost for collected liters in the year 2019 and the cost for the maximum capacity of the bank for that year of activity. Analyzing each cost item that determines the charge of donor human milk (DHM), the highest costs are the salaries of medical and paramedical staff, and then the costs related to transporting. If the HMB works at maximum capacity and manages a greater number of liters of milk, this can represent an important saving. Conversely, the price of consumables is modest (i.e., the price of a single-use kit for breast pumps was € 0.22 per unit). CONCLUSION The costs for a liter of DHM are quite high, but they must be related to the benefits, especially for preterm infants. Comparing the cost for collected liters in 2019 and the costs for the 2019 maximum capacity of the HMB, we calculated how much fixed costs of collection and distribution of DHM can be reduced, by increasing the volume of milk collected. To the best of our knowledge, this is the first complete cost analysis for an Italian Milk Bank. A thorough analysis could help to abate fixed costs and reduce the cost of a liter of DHM. The centralization of DHM can allow savings, rather than creating small HMBs scattered throughout the territory that would operate with lower milk volumes.
Collapse
Affiliation(s)
- Guglielmo Salvatori
- grid.414125.70000 0001 0727 6809Neonatal Intensive Care Unit and Human Milk Bank, Medical and Surgical Department of Fetus - Newborn – Infant, “Bambino Gesù” Children’s Hospital IRCCS, Rome, Italy
| | - Domenico Umberto De Rose
- grid.414125.70000 0001 0727 6809Neonatal Intensive Care Unit and Human Milk Bank, Medical and Surgical Department of Fetus - Newborn – Infant, “Bambino Gesù” Children’s Hospital IRCCS, Rome, Italy
| | - Maria Clemente
- Neonatal Intensive Care Unit and Human Milk Bank, Department of Life and Reproductive Sciences, Verona Hospital, Verona, Italy
| | - Cristina Gentili
- grid.414125.70000 0001 0727 6809Finance Control, Internal Control, “Bambino Gesù” Children’s Hospital IRCCS, Rome, Italy
| | - Giovanni Paride Verardi
- grid.414125.70000 0001 0727 6809Finance Control, Internal Control, “Bambino Gesù” Children’s Hospital IRCCS, Rome, Italy
| | - Patrizia Amadio
- grid.414125.70000 0001 0727 6809Neonatal Intensive Care Unit and Human Milk Bank, Medical and Surgical Department of Fetus - Newborn – Infant, “Bambino Gesù” Children’s Hospital IRCCS, Rome, Italy
| | - Maria Paola Reposi
- grid.414125.70000 0001 0727 6809Neonatal Intensive Care Unit and Human Milk Bank, Medical and Surgical Department of Fetus - Newborn – Infant, “Bambino Gesù” Children’s Hospital IRCCS, Rome, Italy
| | - Pietro Bagolan
- grid.414125.70000 0001 0727 6809Neonatal Surgery Unit, Medical and Surgical Department of Fetus - Newborn – Infant, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Andrea Dotta
- grid.414125.70000 0001 0727 6809Neonatal Intensive Care Unit and Human Milk Bank, Medical and Surgical Department of Fetus - Newborn – Infant, “Bambino Gesù” Children’s Hospital IRCCS, Rome, Italy
| |
Collapse
|
7
|
Benkerroum N, Ismail A. Human Breast Milk Contamination with Aflatoxins, Impact on Children's Health, and Possible Control Means: A Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:16792. [PMID: 36554670 PMCID: PMC9779431 DOI: 10.3390/ijerph192416792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 06/17/2023]
Abstract
Aflatoxins are natural toxicants produced mainly by species of the Aspergillus genus, which contaminate virtually all feeds and foods. Apart from their deleterious health effects on humans and animals, they can be secreted unmodified or carried over into the milk of lactating females, thereby posing health risks to suckling babies. Aflatoxin M1 (AFM1) is the major and most toxic aflatoxin type after aflatoxin B1 (AFB1). It contaminates human breast milk upon direct ingestion from dairy products or by carry-over from the parent molecule (AFB1), which is hydroxylated in the liver and possibly in the mammary glands by cytochrome oxidase enzymes and then excreted into breast milk as AFM1 during lactation via the mammary alveolar epithelial cells. This puts suckling infants and children fed on this milk at a high risk, especially that their detoxifying activities are still weak at this age essentially due to immature liver as the main organ responsible for the detoxification of xenobiotics. The occurrence of AFM1 at toxic levels in human breast milk and associated health conditions in nursing children is well documented, with developing countries being the most affected. Different studies have demonstrated that contamination of human breast milk with AFM1 represents a real public health issue, which should be promptly and properly addressed to reduce its incidence. To this end, different actions have been suggested, including a wider and proper implementation of regulatory measures, not only for breast milk but also for foods and feeds as the upstream sources for breast milk contamination with AFM1. The promotion of awareness of lactating mothers through the organization of training sessions and mass media disclosures before and after parturition is of a paramount importance for the success of any action. This is especially relevant that there are no possible control measures to ensure compliance of lactating mothers to specific regulatory measures, which can yet be appropriate for the expansion of breast milk banks in industrialized countries and emergence of breast milk sellers. This review attempted to revisit the public health issues raised by mother milk contamination with AFM1, which remains undermined despite the numerous relevant publications highlighting the needs to tackle its incidence as a protective measure for the children physical and mental health.
Collapse
Affiliation(s)
- Noreddine Benkerroum
- Expertise Aliments Santé, Food Health Consultancy, 7450 Dollier Str., Montréal, QC H1S 2J6, Canada
| | - Amir Ismail
- Institute of Food Science and Nutrition, Bahauddin Zakariya University, Multan 60000, Pakistan
| |
Collapse
|
8
|
Ren Z, Mo W, Yang L, Wang J, Zhang Q, Zhong Z, Wei W, Liu Z, Wu Z, Yao Y, Yang J. Cord blood antimicrobial peptide LL37 levels in preterm neonates and association with preterm complications. Ital J Pediatr 2022; 48:111. [PMID: 35804392 PMCID: PMC9270758 DOI: 10.1186/s13052-022-01295-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/06/2022] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Cathelicidin/LL-37 plays a significant role in the human immune defense reaction. Preterm human immature organs being exposed to inflammation-induced injury was the critical denominator leading to the common preterm associated complications. Previous study showed LL37 concentration in preterm neonates was lower in tracheal aspirates and breast milk as compared to term infants. An adults study showed decreased LL-37 levels was a risk factor for patients in developing severe chronic obstructive pulmonary disease (COPD). However, little is known about the regulation of human cord blood LL37 in preterm neonates and the association with preterm complications. This study was designed to investigate the concentration of LL37 in cord blood of preterm infants and correlation with preterm complications. METHODS Singleton infants born in June 2017 to August 2021 in the study hospital were enrolled. Maternal and neonatal clinical characteristics were collected. LL37 levels, pro-inflammatory factor interleukin-6 (IL-6) and tumor necrosis factor-a (TNF-a) in cord blood and LL37 levels in serum 48-72 hours after birth were measured by enzyme-linked immunosorbent assay. The serum level of LL37 in preterm and term neonates were compared, the perinatal factors possibly affecting the LL37 levels were investigated and the relationship between LL37 level and preterm outcomes were analyzed. RESULTS Cord blood LL37 levels in preterm infants were lower than that in term neonates. Cord blood LL37 level was positively correlated with gestational age in preterm. Prenatal steroid administration in preterm neonates decreased cord blood LL37 level. LL37 level was obviously lower in patients with bronchopulmonary dysplasia (BPD). Multiple line regression analysis showed higher LL37 level in cord blood was an independent protective factor for BPD. The concentration of pro-inflammatory factor IL-6 was negatively correlated with LL37. CONCLUSION Cord blood LL37 levels increased during gestation and decreased after perinatal steroid usage. Very preterm infants who displayed higher cord blood LL37 level had reduced risk of developing BPD. Regulation of pro-inflammatory cytokine IL-6 may be associated with the protective effect of LL37 on BPD.
Collapse
Affiliation(s)
- Zhuxiao Ren
- Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China
| | - Wenhui Mo
- Department of Neonatology, Foshan fosun chancheng hospital, Foshan, China.
| | - Liling Yang
- Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China
| | - Jianlan Wang
- Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China
| | - Qi Zhang
- Clinical Genetic Center, Guangdong Women and Children Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhicheng Zhong
- Clinical Genetic Center, Guangdong Women and Children Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wei Wei
- Guangdong Cord Blood Bank, Guangzhou, China
| | | | - Zhiping Wu
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Center for Cell Regeneration and Biological Therapies, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yao Yao
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Center for Cell Regeneration and Biological Therapies, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Jie Yang
- Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China. .,Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
9
|
Hatmal MM, Al-Hatamleh MAI, Olaimat AN, Alshaer W, Hasan H, Albakri KA, Alkhafaji E, Issa NN, Al-Holy MA, Abderrahman SM, Abdallah AM, Mohamud R. Immunomodulatory Properties of Human Breast Milk: MicroRNA Contents and Potential Epigenetic Effects. Biomedicines 2022; 10:1219. [PMID: 35740242 PMCID: PMC9219990 DOI: 10.3390/biomedicines10061219] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023] Open
Abstract
Infants who are exclusively breastfed in the first six months of age receive adequate nutrients, achieving optimal immune protection and growth. In addition to the known nutritional components of human breast milk (HBM), i.e., water, carbohydrates, fats and proteins, it is also a rich source of microRNAs, which impact epigenetic mechanisms. This comprehensive work presents an up-to-date overview of the immunomodulatory constituents of HBM, highlighting its content of circulating microRNAs. The epigenetic effects of HBM are discussed, especially those regulated by miRNAs. HBM contains more than 1400 microRNAs. The majority of these microRNAs originate from the lactating gland and are based on the remodeling of cells in the gland during breastfeeding. These miRNAs can affect epigenetic patterns by several mechanisms, including DNA methylation, histone modifications and RNA regulation, which could ultimately result in alterations in gene expressions. Therefore, the unique microRNA profile of HBM, including exosomal microRNAs, is implicated in the regulation of the genes responsible for a variety of immunological and physiological functions, such as FTO, INS, IGF1, NRF2, GLUT1 and FOXP3 genes. Hence, studying the HBM miRNA composition is important for improving the nutritional approaches for pregnancy and infant's early life and preventing diseases that could occur in the future. Interestingly, the composition of miRNAs in HBM is affected by multiple factors, including diet, environmental and genetic factors.
Collapse
Affiliation(s)
- Ma’mon M. Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Mohammad A. I. Al-Hatamleh
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Malaysia;
| | - Amin N. Olaimat
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan; (A.N.O.); (M.A.A.-H.)
| | - Walhan Alshaer
- Cell Therapy Center (CTC), The University of Jordan, Amman 11942, Jordan;
| | - Hanan Hasan
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan;
| | - Khaled A. Albakri
- Faculty of Medicine, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Enas Alkhafaji
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, The University of Jordan, Amman 11942, Jordan;
| | - Nada N. Issa
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Murad A. Al-Holy
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan; (A.N.O.); (M.A.A.-H.)
| | - Salim M. Abderrahman
- Department of Biology and Biotechnology, Faculty of Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Atiyeh M. Abdallah
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar;
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Malaysia;
| |
Collapse
|
10
|
Lopes BS, Hanafiah A, Nachimuthu R, Muthupandian S, Md Nesran ZN, Patil S. The Role of Antimicrobial Peptides as Antimicrobial and Antibiofilm Agents in Tackling the Silent Pandemic of Antimicrobial Resistance. Molecules 2022; 27:molecules27092995. [PMID: 35566343 PMCID: PMC9105241 DOI: 10.3390/molecules27092995] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 01/11/2023] Open
Abstract
Just over a million people died globally in 2019 due to antibiotic resistance caused by ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species). The World Health Organization (WHO) also lists antibiotic-resistant Campylobacter and Helicobacter as bacteria that pose the greatest threat to human health. As it is becoming increasingly difficult to discover new antibiotics, new alternatives are needed to solve the crisis of antimicrobial resistance (AMR). Bacteria commonly found in complex communities enclosed within self-produced matrices called biofilms are difficult to eradicate and develop increased stress and antimicrobial tolerance. This review summarises the role of antimicrobial peptides (AMPs) in combating the silent pandemic of AMR and their application in clinical medicine, focusing on both the advantages and disadvantages of AMPs as antibiofilm agents. It is known that many AMPs display broad-spectrum antimicrobial activities, but in a variety of organisms AMPs are not stable (short half-life) or have some toxic side effects. Hence, it is also important to develop new AMP analogues for their potential use as drug candidates. The use of one health approach along with developing novel therapies using phages and breakthroughs in novel antimicrobial peptide synthesis can help us in tackling the problem of AMR.
Collapse
Affiliation(s)
- Bruno S. Lopes
- Department of Medical Microbiology, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
- Correspondence: (B.S.L.); (A.H.)
| | - Alfizah Hanafiah
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
- Correspondence: (B.S.L.); (A.H.)
| | - Ramesh Nachimuthu
- Antibiotic Resistance and Phage Therapy Laboratory, Department of Biomedical Sciences, Vellore Institute of Technology, School of Bioscience and Technology, Vellore 632014, India;
| | - Saravanan Muthupandian
- AMR and Nanotherapeutics Laboratory, Department of Pharmacology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College, Chennai 600077, India;
| | - Zarith Nameyrra Md Nesran
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Sandip Patil
- Department of Hematology and Oncology, Shenzhen Children’s Hospital, Shenzhen 518038, China;
| |
Collapse
|
11
|
Dong D, Ru X, Huang X, Sang T, Li S, Wang Y, Feng Q. A prospective cohort study on lactation status and breastfeeding challenges in mothers giving birth to preterm infants. Int Breastfeed J 2022; 17:6. [PMID: 35012631 PMCID: PMC8751123 DOI: 10.1186/s13006-021-00447-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/26/2021] [Indexed: 11/18/2022] Open
Abstract
Background Mothers of preterm infants face many challenges in breastfeeding, especially regarding lactation. This study aimed to investigate the lactation status and challenges in breastfeeding faced by preterm infants’ mothers. Methods We approached 124 mothers who gave birth to preterm infants between 26 May and 31 October 2018 in a tertiary hospital in China. Lactation status and challenges in breastfeeding on day 7 postpartum, at discharge of infants, 2 weeks post-discharge, and 3 months of corrected age were collected using questionnaires. The area under the receiver operating characteristic (ROC) curve for expressed milk volume on day 7 postpartum for predicting expressed milk volume ≥ 300 mL/d at discharge was calculated. Logistic regression analyses were performed to identify factors associated with delayed lactogenesis II onset and continuation of breastfeeding at 3 months of corrected age. Results Seventy mothers were enrolled, and 51.4% had delayed lactogenesis II. Multivariate logistic regression analysis revealed that older maternal age (aOR = 1.19; 95% CI: 1.01, 1.40) and first live birth (aOR = 4.81; 95% CI 1.43, 16.18) were significant independent predictors of delayed lactogenesis II. Mothers with delayed lactogenesis II had significantly lower expressed milk volume (day 7 postpartum: 160.0 mL vs. 300.0 mL, U = 328.50, p = 0.001; at discharge: 425.0 mL vs. 612.5 mL, U = 372.00, p = 0.005), with a lower proportion of exclusive breastfeeding in their infants (at discharge: 33.3% vs. 69.8%, χ2 = 12.39, df = 1, p < 0.001; 3 months of corrected age: 17.8% vs. 52.8%, χ2 = 11.03, df = 1, p = 0.001). The ROC showed that expressed milk volume > 190 mL/d on day 7 postpartum significantly predicted expressed milk volume ≥ 300 mL/d at discharge. Insufficient human milk was the main reason for breastfeeding discontinuation at 3 months of corrected age. Twins were less likely to continue breastfeeding at 3 months of corrected age (aOR = 0.27; 95% CI 0.09, 0.86). In singleton infants, mother’s own milk ≥50% of total milk uptake at 2 weeks post-discharge (aOR = 32.66; 95% CI 3.00, 355.25) was an independent predictor of continuous breastfeeding at 3 months of corrected age. Feeding complications in infants, poor breastfeeding technique, and low milk output are the main challenges in breastfeeding. Conclusion Interventions to improve early postpartum lactation and breastfeeding techniques may increase breastfeeding adoption in mothers of preterm infants. Supplementary Information The online version contains supplementary material available at 10.1186/s13006-021-00447-4.
Collapse
Affiliation(s)
- Dingding Dong
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'anmen Street, Xicheng District, Beijing, China
| | - Xifang Ru
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'anmen Street, Xicheng District, Beijing, China
| | - Xiaofang Huang
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'anmen Street, Xicheng District, Beijing, China
| | - Tian Sang
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'anmen Street, Xicheng District, Beijing, China
| | - Shan Li
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'anmen Street, Xicheng District, Beijing, China
| | - Ying Wang
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'anmen Street, Xicheng District, Beijing, China
| | - Qi Feng
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'anmen Street, Xicheng District, Beijing, China.
| |
Collapse
|
12
|
Faust KB, Moser K, Bartels M, Fortmann I, Hanke K, Wieg C, Stichtenoth G, Göpel W, Herting E, Härtel C. Lactoferrin and Human Neutrophil Protein (HNP) 1-3 Levels During the Neonatal Period in Preterm Infants. Front Pediatr 2022; 10:909176. [PMID: 35967550 PMCID: PMC9364083 DOI: 10.3389/fped.2022.909176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Antimicrobial polypeptides (APPs) are part of the innate immune system, but their specific role in the context of preterm birth is not yet understood. The aim of this investigation was to determine the systemic expression of APPs, i.e., lactoferrin (LF) and human neutrophil protein (HNP) 1-3 in preterm infants in the period of highest vulnerability for infection and to correlate these biomarkers with short-term outcome. We therefore conducted a prospective two-center study including plasma samples of 278 preterm infants and 78 corresponding mothers. APP levels were analyzed on day 1, 3, 7, and 21 of life via enzyme-linked immunosorbent assay (ELISA). The levels of LF and HNP1-3 remained stable during the first 21 days of life and were not influenced by maternal levels. Elevated APP levels were found at day 1 in infants born to mothers with amniotic infection syndrome (AIS vs. no AIS, mean ± SD in ng/ml: LF 199.8 ± 300 vs. 124.1 ± 216.8, HNP 1-3 16,819 ± 36,124 vs. 8,701 ± 11,840; p = 0.021, n = 179). We found no elevated levels of APPs before the onset of sepsis episodes or in association with other short-term outcomes that are in part mediated by inflammation such as necrotizing enterocolitis (NEC) or retinopathy of prematurity (ROP). Interestingly, infants developing bronchopulmonary dysplasia (BPD) showed higher levels of HNP1-3 on day 21 than infants without BPD (13,473 ± 16,135 vs. 8,388 ± 15,938, n = 111, p = 0.008). In infants born without amniotic infection, levels of the measured APPs correlated with gestational age and birth weight. In our longitudinal study, systemic levels of LF and HNP 1-3 were not associated with postnatal infection and adverse short-term outcomes in preterm infants.
Collapse
Affiliation(s)
- Kirstin B Faust
- Department of Paediatrics, University of Lübeck, Lübeck, Germany
| | - Katja Moser
- Department of Neonatology and Pediatric Intensive Care, Hospital Aschaffenburg-Alzenau, Aschaffenburg, Germany
| | - Maren Bartels
- Department of Paediatrics, University of Lübeck, Lübeck, Germany
| | - Ingmar Fortmann
- Department of Paediatrics, University of Lübeck, Lübeck, Germany
| | - Kathrin Hanke
- Department of Paediatrics, University of Lübeck, Lübeck, Germany
| | - Christian Wieg
- Department of Neonatology and Pediatric Intensive Care, Hospital Aschaffenburg-Alzenau, Aschaffenburg, Germany
| | | | - Wolfgang Göpel
- Department of Paediatrics, University of Lübeck, Lübeck, Germany
| | - Egbert Herting
- Department of Paediatrics, University of Lübeck, Lübeck, Germany
| | - Christoph Härtel
- Department of Pediatrics, University of Würzburg, Würzburg, Germany
| |
Collapse
|
13
|
Healy DB, Ryan CA, Ross RP, Stanton C, Dempsey EM. Clinical implications of preterm infant gut microbiome development. Nat Microbiol 2022; 7:22-33. [PMID: 34949830 DOI: 10.1038/s41564-021-01025-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 11/12/2021] [Indexed: 12/12/2022]
Abstract
Perturbations to the infant gut microbiome during the first weeks to months of life affect growth, development and health. In particular, assembly of an altered intestinal microbiota during infant development results in an increased risk of immune and metabolic diseases that can persist into childhood and potentially into adulthood. Most research into gut microbiome development has focused on full-term babies, but health-related outcomes are also important for preterm babies. The systemic physiological immaturity of very preterm gestation babies (born earlier than 32 weeks gestation) results in numerous other microbiome-organ interactions, the mechanisms of which have yet to be fully elucidated or in some cases even considered. In this Perspective, we compare assembly of the intestinal microbiome in preterm and term infants. We focus in particular on the clinical implications of preterm infant gut microbiome composition and discuss the prospects for microbiome diagnostics and interventions to improve the health of preterm babies.
Collapse
Affiliation(s)
- David B Healy
- APC Microbiome Ireland, University College Cork, Cork, Ireland. .,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.
| | - C Anthony Ryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | - Eugene M Dempsey
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.,INFANT Research Centre, Cork University Hospital, Cork, Ireland
| |
Collapse
|
14
|
Cieślik M, Bagińska N, Górski A, Jończyk-Matysiak E. Human β-Defensin 2 and Its Postulated Role in Modulation of the Immune Response. Cells 2021; 10:cells10112991. [PMID: 34831214 PMCID: PMC8616480 DOI: 10.3390/cells10112991] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/28/2021] [Accepted: 10/30/2021] [Indexed: 12/17/2022] Open
Abstract
Studies described so far suggest that human β-defensin 2 is an important protein of innate immune response which provides protection for the human organism against invading pathogens of bacterial, viral, fungal, as well as parasitical origin. Its pivotal role in enhancing immunity was proved in infants. It may also be considered a marker of inflammation. Its therapeutic administration has been suggested for maintenance of the balance of systemic homeostasis based on the appropriate composition of the microbiota. It has been suggested that it may be an important therapeutic tool for modulating the response of the immune system in many inflammatory diseases, offering new treatment modalities. For this reason, its properties and role in the human body discussed in this review should be studied in more detail.
Collapse
Affiliation(s)
- Martyna Cieślik
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland; (M.C.); (N.B.); (E.J.-M.)
| | - Natalia Bagińska
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland; (M.C.); (N.B.); (E.J.-M.)
| | - Andrzej Górski
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland; (M.C.); (N.B.); (E.J.-M.)
- Phage Therapy Unit, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland
- Infant Jesus Hospital, The Medical University of Warsaw, 02-006 Warsaw, Poland
- Correspondence:
| | - Ewa Jończyk-Matysiak
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland; (M.C.); (N.B.); (E.J.-M.)
| |
Collapse
|
15
|
Milk Osteopontin for Gut, Immunity and Brain Development in Preterm Pigs. Nutrients 2021; 13:nu13082675. [PMID: 34444835 PMCID: PMC8400468 DOI: 10.3390/nu13082675] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 11/29/2022] Open
Abstract
Deficient levels of milk osteopontin (OPN) in infant formula may partly account for developmental differences between infants fed formula or maternal milk. We hypothesized that a milk diet supplemented with bovine milk OPN improves gut, immunity and brain development and tested this in a preterm pig model. Preterm pigs delivered by cesarean section (90% gestation) were fed raw bovine milk (CON, n = 19) or the same diet supplemented with a physiologically relevant dose of OPN (46 mg/(kg·d), n = 16). Endpoints related to clinical outcomes, systemic immunity and neurocognitive development were assessed during the study and gut tissues were collected at Day 19. Growth pattern, early motor development and most systemic immune parameters were similar between OPN and CON pigs. The OPN pigs had higher villus-to-crypt ratios than CON pigs and higher monocyte and lymphocyte counts on Day 8. Gut digestive and absorptive functions and cognitive performance (T-maze test) were similar between OPN and CON pigs. In conclusion, dietary supplementation with OPN above basal bovine milk levels induced minor improvements in gut structure and systemic immunity without any effects on cognitive performance. The minimal levels of OPN in infant formula to secure optimal adaptation in the immediate neonatal period remain to be determined.
Collapse
|
16
|
Sangild PT, Vonderohe C, Melendez Hebib V, Burrin DG. Potential Benefits of Bovine Colostrum in Pediatric Nutrition and Health. Nutrients 2021; 13:nu13082551. [PMID: 34444709 PMCID: PMC8402036 DOI: 10.3390/nu13082551] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/02/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022] Open
Abstract
Bovine colostrum (BC), the first milk produced from cows after parturition, is increasingly used as a nutritional supplement to promote gut function and health in other species, including humans. The high levels of whey and casein proteins, immunoglobulins (Igs), and other milk bioactives in BC are adapted to meet the needs of newborn calves. However, BC supplementation may improve health outcomes across other species, especially when immune and gut functions are immature in early life. We provide a review of BC composition and its effects in infants and children in health and selected diseases (diarrhea, infection, growth-failure, preterm birth, necrotizing enterocolitis (NEC), short-bowel syndrome, and mucositis). Human trials and animal studies (mainly in piglets) are reviewed to assess the scientific evidence of whether BC is a safe and effective antimicrobial and immunomodulatory nutritional supplement that reduces clinical complications related to preterm birth, infections, and gut disorders. Studies in infants and animals suggest that BC should be supplemented at an optimal age, time, and level to be both safe and effective. Exclusive BC feeding is not recommended for infants because of nutritional imbalances relative to human milk. On the other hand, adverse effects, including allergies and intolerance, appear unlikely when BC is provided as a supplement within normal nutrition guidelines for infants and children. Larger clinical trials in infant populations are needed to provide more evidence of health benefits when patients are supplemented with BC in addition to human milk or formula. Igs and other bioactive factors in BC may work in synergy, making it critical to preserve bioactivity with gentle processing and pasteurization methods. BC has the potential to become a safe and effective nutritional supplement for several pediatric subpopulations.
Collapse
Affiliation(s)
- Per Torp Sangild
- Comparative Pediatrics & Nutrition, University of Copenhagen, DK-1870 Copenhagen, Denmark;
- Department of Neonatology, Rigshospitalet, DK-1870 Copenhagen, Denmark
- Department of Pediatrics, Odense University Hospital, DK-5000 Odense, Denmark
| | - Caitlin Vonderohe
- USDA-ARS Children’s Nutrition Research Center, Pediatrics, Gastroenterology & Nutrition, Baylor College of Medicine, Houston, TX 77030, USA; (C.V.); (V.M.H.)
| | - Valeria Melendez Hebib
- USDA-ARS Children’s Nutrition Research Center, Pediatrics, Gastroenterology & Nutrition, Baylor College of Medicine, Houston, TX 77030, USA; (C.V.); (V.M.H.)
| | - Douglas G. Burrin
- USDA-ARS Children’s Nutrition Research Center, Pediatrics, Gastroenterology & Nutrition, Baylor College of Medicine, Houston, TX 77030, USA; (C.V.); (V.M.H.)
- Correspondence: ; Tel.: +1-713-798-7049
| |
Collapse
|
17
|
Wagner C, Torow N, Hornef MW, Lelouard H. Spatial and temporal key steps in early-life intestinal immune system development and education. FEBS J 2021; 289:4731-4757. [PMID: 34076962 DOI: 10.1111/febs.16047] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/15/2021] [Accepted: 06/01/2021] [Indexed: 12/15/2022]
Abstract
Education of our intestinal immune system early in life strongly influences adult health. This education strongly relies on series of events that must occur in well-defined time windows. From initial colonization by maternal-derived microbiota during delivery to dietary changes from mother's milk to solid foods at weaning, these early-life events have indeed long-standing consequences on our immunity, facilitating tolerance to environmental exposures or, on the contrary, increasing the risk of developing noncommunicable diseases such as allergies, asthma, obesity, and inflammatory bowel diseases. In this review, we provide an outline of the recent advances in our understanding of these events and how they are mechanistically related to intestinal immunity development and education. First, we review the susceptibility of neonates to infections and inflammatory diseases, related to their immune system and microbiota changes. Then, we highlight the maternal factors involved in protection and education of the mucosal immune system of the offspring, the role of the microbiota, and the nature of neonatal immune system until weaning. We also present how the development of some immune responses is intertwined in temporal and spatial windows of opportunity. Finally, we discuss pending questions regarding the neonate particular immune status and the activation of the intestinal immune system at weaning.
Collapse
Affiliation(s)
- Camille Wagner
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Natalia Torow
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Mathias W Hornef
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | | |
Collapse
|
18
|
Cai J, Li X, Wang X, Jiang C, Shen D, Cui X, Xie K, Ji C, Cao Y. A human β-casein-derived peptide BCCY-1 modulates the innate immune response. Food Chem 2021; 348:129111. [PMID: 33516994 DOI: 10.1016/j.foodchem.2021.129111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 01/11/2021] [Accepted: 01/11/2021] [Indexed: 12/21/2022]
Abstract
In this study, we report a novel peptide corresponding to the sequence of human β-casein (named BCCY-1), which was identified in our previous peptidome analysis of human milk and has great immunomodulatory activity. The results revealed that peptide BCCY-1, but not the scrambled version, enhanced monocyte migration without obvious toxicities. This selective effect was mediated via increased production of chemokines by peptide stimulated monocytes. Moreover, BCCY-1 exerted its modulatory effects by activating nuclear factor (NF)-κB and mitogen-activated protein kinase (MAPK) signaling. The abundances of peptide BCCY-1 and the peptides partially encompassing its fragment were found to be lower in preterm milk than in term milk. Our study may lead to new insights into the immunoregulatory effects of casein-derived peptides and facilitate the discovery of novel peptide-based food and pharmaceutical products.
Collapse
Affiliation(s)
- Jinyang Cai
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Xin Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Xing Wang
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China
| | - Chengfei Jiang
- Department of Pathology, Nanjing Medical University, Nanjing 210029, China
| | - Dan Shen
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China
| | - Xianwei Cui
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China
| | - Kaipeng Xie
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China
| | - Chenbo Ji
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China.
| | - Yan Cao
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China.
| |
Collapse
|
19
|
Nadimpalli ML, Bourke CD, Robertson RC, Delarocque-Astagneau E, Manges AR, Pickering AJ. Can breastfeeding protect against antimicrobial resistance? BMC Med 2020; 18:392. [PMID: 33317529 PMCID: PMC7737306 DOI: 10.1186/s12916-020-01862-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 11/19/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND The proportion of infections among young children that are antimicrobial-resistant is increasing across the globe. Newborns may be colonized with enteric antimicrobial-resistant pathogens early in life, which is a risk factor for infection-related morbidity and mortality. Breastfeeding is actively promoted worldwide for its beneficial impacts on newborn health and gut health. However, the role of breastfeeding and human milk components in mitigating young children's carriage of antimicrobial-resistant pathogens and antibiotic resistance genes has not been comprehensively explored. MAIN BODY Here, we review how the act of breastfeeding, early breastfeeding, and/or human milk components, such as the milk microbiota, secretory IgA, human milk oligosaccharides, antimicrobial peptides, and microRNA -bearing extracellular vesicles, could play a role in preventing the establishment of antimicrobial-resistant pathogens in young children's developing gut microbiomes. We describe findings from recent human studies that support this concept. CONCLUSION Given the projected rise in global morbidity and mortality that will stem from antimicrobial-resistant infections, identifying behavioral or nutritional interventions that could decrease children's susceptibility to colonization with antimicrobial-resistant pathogens may be one strategy for protecting their health. We suggest that breastfeeding and human milk supplements deserve greater attention as potential preventive measures in the global effort to combat antimicrobial resistance, particularly in low- and middle-income settings.
Collapse
Affiliation(s)
- Maya L Nadimpalli
- Department of Civil and Environmental Engineering, Tufts University, Science & Engineering Complex, Anderson Hall, Room 204, 200 College Avenue, Medford, MA, USA. .,Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance (Levy CIMAR), Tufts University, Boston, MA, USA.
| | - Claire D Bourke
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, E1 2AT, UK.,Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Ruairi C Robertson
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, E1 2AT, UK
| | - Elisabeth Delarocque-Astagneau
- Université Paris-Saclay, UVSQ, Inserm, CESP, Team Anti-infective Evasion and Pharmacoepidemiology, 78180 Montigny, France.,AP-HP, GHU Paris Saclay University, Raymond Poincaré Hospital, Epidemiology and Public Health Department, 92380 Garches, France
| | - Amee R Manges
- School of Population and Public Health, The University of British Columbia, Vancouver, BC, Canada.,British Columbia Centre for Disease Control, Vancouver, BC, Canada
| | - Amy J Pickering
- Department of Civil and Environmental Engineering, Tufts University, Science & Engineering Complex, Anderson Hall, Room 204, 200 College Avenue, Medford, MA, USA.,Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance (Levy CIMAR), Tufts University, Boston, MA, USA
| |
Collapse
|
20
|
Vertical transmission of gut microbiota: Points of action of environmental factors influencing brain development. Neurosci Res 2020; 168:83-94. [PMID: 33309866 DOI: 10.1016/j.neures.2020.11.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/13/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022]
Abstract
Environmental factors in early life interact with genetics to exert a long-lasting and broad influence on health and disease. There has been a marked growth in the number of environmental factors studied in association with neurodevelopmental disorders. Colonization of the gut microbiota in the offspring uses the maternal resident flora as a primary source of bacteria during perinatal periods. Several lines of evidence have shown that various environmental factors including the mode of delivery, exposure to antibiotics, infection, stress, diet, quality of breast milk, and type of infant-feeding during the perinatal periods can perturb the gut microbiota colonization in the offspring, finally leading to disturbances in brain development. This study proposes that the gut microbiota seeded primarily by maternal microbiota, and the postnatal colonization of the microbiota in the offspring can be critical action points of environmental factors when deciphering the mechanisms of actions of environmental factors in brain development. This research reviews the inheritance and colonization of the microbiota during early life and the potential actions of the environmental factors influencing brain development in the offspring by modulating the vertical transmission of gut microbiota.
Collapse
|
21
|
Gao X, Li Y, Olin AB, Nguyen DN. Fortification With Bovine Colostrum Enhances Antibacterial Activity of Human Milk. JPEN J Parenter Enteral Nutr 2020; 45:1417-1424. [PMID: 33305396 PMCID: PMC8647157 DOI: 10.1002/jpen.2060] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/24/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023]
Abstract
Objectives Human milk (HM) is the optimal diet for neonates, but it does not provide enough nutrients for preterm infants. HM fortifiers based on highly processed mature bovine milk (BMFs) are routinely used for preterm infants despite risks of causing gut dysfunction and systemic infection. Gently‐processed bovine colostrum as a fortifier (BCF) may better protect against infection and inflammation. We hypothesized that BCF‐fortified HM has enhanced antimicrobial activity against pathogens that commonly cause neonatal sepsis, relative to BMF‐fortified HM. Methods Holder‐pasteurized HM samples (10 mothers) were aliquoted into 3 fractions: unfortified HM and HM fortified with either BMF or BCF. The samples were analyzed for pH, lactoferrin concentrations, and antimicrobial activities against Staphylococcus epidermidis, Escherichia coli, and Enterococcus faecalis. Results HM+BCF had a lower pH and higher lactoferrin levels than HM+BMF, with HM being intermediate. Relative to infant formula, HM decreased the growth of S epidermidis, E coli, and E faecalis, with no difference between preterm and term HM. Addition of BMF abolished the antimicrobial effect of HM against S epidermidis and E faecalis but not E coli. By contrast, addition of BCF into HM enhanced antimicrobial activity against S epidermidis and E coli, relative to unfortified HM. HM+BCF was superior to HM+BMF in inhibiting growth of all tested bacteria. Conclusion BMF fortification decreased whereas BCF fortification enhanced in vitro antimicrobial activity of HM. This effect may partly be derived from the high levels of antimicrobial factors found in BCF, including lactoferrin. BCF may be a better fortifier than BMF for preterm infants.
Collapse
Affiliation(s)
- Xiaoyan Gao
- Department of Neonatology, Affiliated Foshan Maternity Child Healthcare Hospital, Southern Medical University, Foshan, China.,Department of Neonatology, Foshan Maternity Child Healthcare Hospital, Foshan, China.,Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yanqi Li
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Bille Olin
- National Human Milk Bank, Hvidovre Hospital, Copenhagen, Denmark
| | - Duc Ninh Nguyen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Two-Component Signal Transduction Systems in the Human Pathogen Streptococcus agalactiae. Infect Immun 2020; 88:IAI.00931-19. [PMID: 31988177 DOI: 10.1128/iai.00931-19] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Streptococcus agalactiae (group B Streptococcus [GBS]) is an important cause of invasive infection in newborns, maternal women, and older individuals with underlying chronic illnesses. GBS has many mechanisms to adapt and survive in its host, and these mechanisms are often controlled via two-component signal transduction systems. In GBS, more than 20 distinct two-component systems (TCSs) have been classified to date, consisting of canonical TCSs as well as orphan and atypical sensors and regulators. These signal transducing systems are necessary for metabolic regulation, resistance to antibiotics and antimicrobials, pathogenesis, and adhesion to the mucosal surfaces to colonize the host. This minireview discusses the structures of these TCSs in GBS as well as how selected systems regulate essential cellular processes such as survival and colonization. GBS contains almost double the number of TCSs compared to the closely related Streptococcus pyogenes and Streptococcus pneumoniae, and while research on GBS TCSs has been increasing in recent years, no comprehensive reviews of these TCSs exist, making this review especially relevant.
Collapse
|
23
|
Jahan M, Francis N, Wang B. Milk lactoferrin concentration of primiparous and multiparous sows during lactation. J Dairy Sci 2020; 103:7521-7530. [PMID: 32448579 DOI: 10.3168/jds.2020-18148] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 03/18/2020] [Indexed: 01/21/2023]
Abstract
Lactoferrin (LF), a sialylated iron-binding glycoprotein, has numerous vital physiological functions including immunomodulation and protection against a large group of microorganisms, improving neurodevelopment, health, growth performance, and milk production. Lactoferrin occurs in human milk at a higher concentration compared with bovine milk, but little information is available on LF concentrations in porcine milk and the effects of sow parity on milk LF concentration. The objective of this study was to quantify the LF concentration in porcine milk and to compare that concentration between gilts and sows during lactation. We also investigated the effect of genetic background and litter size of the female pig on the LF concentration of porcine milk. The milk from 30 gilts and 35 sows was collected at 3 stages of lactation, namely colostrum, transition, and mature milk. Standard and experimental samples were analyzed by ultra-high performance liquid chromatography using a diode array UV detector. The following findings were reported: (1) porcine milk contained significant levels of LF with the highest concentration in colostrum, which decreased by ∼62% and ∼67% in transitional and mature milk, respectively; (2) mature gilt milk contained a 22% higher concentration of LF compared with sow milk, which was statistically significant; (3) breed line had an overall significant effect on the LF content of porcine milk; however, when the breed was considered, no significant difference was observed; and (4) LF concentration of porcine milk was not significantly influenced by the litter size. The presence of LF in a higher concentration in porcine milk suggests that LF is an important constituent of pig milk that might contribute to the optimum growth and development of piglets.
Collapse
Affiliation(s)
- M Jahan
- Graham Centre for Agricultural Innovation, Charles Sturt University, Wagga Wagga, NSW 2650, Australia; School of Animal and Veterinary Sciences, Charles Sturt University, Wagga Wagga, NSW 2678, Australia
| | - N Francis
- School of Animal and Veterinary Sciences, Charles Sturt University, Wagga Wagga, NSW 2678, Australia
| | - B Wang
- Graham Centre for Agricultural Innovation, Charles Sturt University, Wagga Wagga, NSW 2650, Australia; School of Animal and Veterinary Sciences, Charles Sturt University, Wagga Wagga, NSW 2678, Australia.
| |
Collapse
|
24
|
Tarnow-Mordi WO, Abdel-Latif ME, Martin A, Pammi M, Robledo K, Manzoni P, Osborn D, Lui K, Keech A, Hague W, Ghadge A, Travadi J, Brown R, Darlow BA, Liley H, Pritchard M, Kochar A, Isaacs D, Gordon A, Askie L, Cruz M, Schindler T, Dixon K, Deshpande G, Tracy M, Schofield D, Austin N, Sinn J, Simes RJ. The effect of lactoferrin supplementation on death or major morbidity in very low birthweight infants (LIFT): a multicentre, double-blind, randomised controlled trial. THE LANCET CHILD & ADOLESCENT HEALTH 2020; 4:444-454. [PMID: 32407710 DOI: 10.1016/s2352-4642(20)30093-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/06/2020] [Accepted: 03/23/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Very low birthweight or preterm infants are at increased risk of adverse outcomes including sepsis, necrotising enterocolitis, and death. We assessed whether supplementing the enteral diet of very low-birthweight infants with lactoferrin, an antimicrobial protein, reduces all-cause mortality or major morbidity. METHODS We did a multicentre, double-blind, pragmatic, randomised superiority trial in 14 Australian and two New Zealand neonatal intensive care units. Infants born weighing less than 1500 g and aged less than 8 days, were eligible and randomly assigned (1:1) using minimising web-based randomisation to receive once daily 200 mg/kg pasteurised bovine lactoferrin supplements or no lactoferrin supplement added to breast or formula milk until 34 weeks' post-menstrual age (or for 2 weeks, if longer), or until discharge from the study hospital if that occurred first. Designated nurses preparing the daily feeds were not masked to group assignment, but other nurses, doctors, parents, caregivers, and investigators were unaware. The primary outcome was survival to hospital discharge or major morbidity (defined as brain injury, necrotising enterocolitis, late-onset sepsis at 36 weeks' post-menstrual age, or retinopathy treated before discharge) assessed in the intention-to-treat population. Safety analyses were by treatment received. We also did a prespecified, PRISMA-compliant meta-analysis, which included this study and other relevant randomised controlled trials, to estimate more precisely the effects of lactoferrin supplementation on late-onset sepsis, necrotising enterocolitis, and survival. This trial is registered with the Australian and New Zealand Clinical Trials Registry, ACTRN12611000247976. FINDINGS Between June 27, 2014, and Sept 1, 2017, we recruited 1542 infants; 771 were assigned to the intervention group and 771 to the control group. One infant who had consent withdrawn before beginning lactoferrin treatment was excluded from analysis. In-hospital death or major morbidity occurred in 162 (21%) of 770 infants in the intervention group and in 170 (22%) of 771 infants in the control group (relative risk [RR] 0·95, 95% CI 0·79-1·14; p=0·60). Three suspected unexpected serious adverse reactions occurred; two in the lactoferrin group, namely unexplained late jaundice and inspissated milk syndrome, but were not attributed to the intervention and one in the control group had fatal inspissated milk syndrome. Our meta-analysis identified 13 trials completed before Feb 18, 2020, including this Article, in 5609 preterm infants. Lactoferrin supplements significantly reduced late-onset sepsis (RR 0·79, 95% CI 0·71-0·88; p<0·0001; I2=58%), but not necrotising enterocolitis or all-cause mortality. INTERPRETATION Lactoferrin supplementation did not improve death or major morbidity in this trial, but might reduce late-onset sepsis, as found in our meta-analysis of over 5000 infants. Future collaborative studies should use products with demonstrated biological activity, be large enough to detect moderate and clinically important effects reliably, and assess greater doses of lactoferrin in infants at increased risk, such as those not exclusively receiving breastmilk or infants of extremely low birthweight. FUNDING Australian National Health and Medical Research Council.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kei Lui
- University of New South Wales, Kensington, NSW, Australia
| | | | - Wendy Hague
- University of Sydney, Sydney, NSW, Australia
| | | | | | | | | | - Helen Liley
- University of Queensland, Brisbane, QLD, Australia
| | | | - Anu Kochar
- University of Adelaide, Adelaide, SA, Australia
| | | | | | - Lisa Askie
- University of Sydney, Sydney, NSW, Australia
| | - Melinda Cruz
- Miracle Babies Foundation, Chipping Norton, NSW, Australia
| | - Tim Schindler
- University of New South Wales, Kensington, NSW, Australia
| | - Kelly Dixon
- University of Queensland, Brisbane, QLD, Australia
| | | | - Mark Tracy
- University of Sydney, Sydney, NSW, Australia
| | | | | | - John Sinn
- University of Sydney, Sydney, NSW, Australia
| | | | | |
Collapse
|
25
|
Ochoa TJ, Mendoza K, Carcamo C, Zegarra J, Bellomo S, Jacobs J, Cossey V. Is Mother's Own Milk Lactoferrin Intake Associated with Reduced Neonatal Sepsis, Necrotizing Enterocolitis, and Death? Neonatology 2020; 117:167-174. [PMID: 32053823 PMCID: PMC7381382 DOI: 10.1159/000505663] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/28/2019] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Lactoferrin (LF) is a protective protein present in milk with anti-infective and immune-modulating properties. OBJECTIVES The aim of this study was to determine the association of maternal LF intake and mother's own milk intake in the first 10 days of life on the prevention of late-onset sepsis (LOS), necrotizing enterocolitis (NEC), or death in the first 8 weeks of life in newborns with a birth weight <2,000 g. METHODS A retrospective cohort study was conducted, with the exposure being the consumption of mother's own LF and mother's own milk in the first 10 days of life, and the outcome being LOS, NEC, or death during days 11 and 56 of life, analyzed by Cox regression. RESULTS Two hundred and ninety-nine infants were enrolled, including 240 with human LF intake information. The average daily human LF intake over days 4-10 of life was 283 mg/kg/day (IQR 114-606 mg/kg/day). The hazard ratio (HR) of mother's own milk LF intake ≥100 mg/kg/day in days 4-10 for LOS, NEC, or death was 0.297 (95% CI 0.156-0.568, p < 0.001); the adjusted HR was 0.752 (95% CI 0.301-1.877, p = 0.541). The adjusted HR of mother's own milk cumulative intake (days 4-10) of 54-344 mL/kg (25-75 quartiles) for LOS, NEC, or death was 0.414 (95% CI 0.196-0.873, p = 0.02). Infants who developed an event (LOS, NEC, or death) had significantly less median daily human LF intake than those that did not (89 vs. 334 mg/kg/day, respectively, p < 0.0001). CONCLUSION Consumption of higher amounts of mother's own milk in the first days of life is associated with less infection, NEC, and death. Early human milk intake should be strongly encouraged in all newborns.
Collapse
Affiliation(s)
- Theresa J Ochoa
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru, .,School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA, .,Doctoral School of Biomedical Sciences, KU Leuven, Leuven, Belgium,
| | - Karina Mendoza
- Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Cesar Carcamo
- Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Jaime Zegarra
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Sicilia Bellomo
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Jan Jacobs
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium.,Department of Clinical Science, Institute of Tropical Medicine, Antwerp, Belgium
| | - Veerle Cossey
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
26
|
Strunk T, Hibbert JE, Doherty D, Nathan E, Simmer K, Patole SK, Trend S, Richmond P, Burgner D, Currie A. Lactoferrin Expression Is Not Associated with Late-Onset Sepsis in Very Preterm Infants. Neonatology 2020; 117:606-611. [PMID: 32862184 DOI: 10.1159/000509404] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/15/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Preterm infants are at a high risk of developing late-onset sepsis (LOS). Lactoferrin is one of the most abundant endogenous antimicrobial proteins expressed in breast milk, stools, and blood, and a candidate for preventive intervention. Large clinical trials have recently investigated whether enteral supplementation with bovine lactoferrin reduces LOS. AIM To characterize lactoferrin levels in preterm infants with and without LOS during the first month of life. METHODS Very preterm and term infants were recruited and serial biosamples collected during the first month of life. Lactoferrin levels were determined by immunoassay in cord blood and peripheral blood on days 1, 7, 14, 21, and 28; in the stools on days 1 and 28; and in the mother's breast milk on days 7 and 21. Furthermore, we assessed the capacity of the peripheral blood to release lactoferrin in response to an in vitro challenge with live Staphylococcus epidermidis, lipopolysaccharide, and fibroblast-stimulating lipopeptide 1. RESULTS Plasma lactoferrin levels were higher in cord blood and day 1 peripheral blood and declined during the first month of life. Plasma lactoferrin levels were similar in term infants and in preterm infants with (n = 32) and without LOS (n = 53). S. epidermidis-induced lactoferrin levels were lower following the sepsis episode. CONCLUSIONS Endogenous lactoferrin expression in preterm infants does not appear to affect their risk of developing LOS. These findings are in line with the lack of benefit recently observed in large trials of enteral supplementation with bovine lactoferrin to prevent LOS.
Collapse
Affiliation(s)
- Tobias Strunk
- Neonatal Directorate, King Edward Memorial Hospital, Perth, Washington, Australia, .,Wesfarmers' Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Washington, Australia, .,School of Medicine, University of Western Australia, Perth, Washington, Australia,
| | - Julie Elizabeth Hibbert
- Wesfarmers' Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Washington, Australia.,School of Medicine, University of Western Australia, Perth, Washington, Australia
| | - Dorota Doherty
- School of Medicine, University of Western Australia, Perth, Washington, Australia
| | - Elizabeth Nathan
- School of Medicine, University of Western Australia, Perth, Washington, Australia
| | - Karen Simmer
- Neonatal Directorate, King Edward Memorial Hospital, Perth, Washington, Australia.,Wesfarmers' Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Washington, Australia.,School of Medicine, University of Western Australia, Perth, Washington, Australia
| | - Sanjay Keshav Patole
- Neonatal Directorate, King Edward Memorial Hospital, Perth, Washington, Australia.,School of Medicine, University of Western Australia, Perth, Washington, Australia
| | - Stephanie Trend
- Wesfarmers' Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Washington, Australia.,Centre for Neuromuscular and Neurological Disorders, Perron Institute for Neurological and Translational Science, University of Western Australia, Perth, Washington, Australia
| | - Peter Richmond
- Wesfarmers' Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Washington, Australia.,School of Medicine, University of Western Australia, Perth, Washington, Australia.,Department of Immunology, Perth Children's Hospital, Perth, Washington, Australia
| | - David Burgner
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew Currie
- Wesfarmers' Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Washington, Australia.,Medical, Molecular and Forensic Sciences, Murdoch University, Perth, Washington, Australia
| |
Collapse
|
27
|
Jackson BA, Gregg BE, Tutor SD, Bermick JR, Stanley KP. Human Milk Retains Important Immunologic Properties After Defatting. JPEN J Parenter Enteral Nutr 2019; 44:904-911. [PMID: 31599047 DOI: 10.1002/jpen.1722] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/09/2019] [Accepted: 09/18/2019] [Indexed: 11/08/2022]
Abstract
BACKGROUND In neonatal chylothorax, thoracic lymphatic drainage is ineffective. The resultant effusions often require drainage, leading to a loss of immune components. Affected infants can be managed with formula or defatted human milk feedings low in long-chain triglycerides to decrease lymph production. We hypothesized that there is no significant difference in the immunological profile or antibacterial effect of full-fat and defatted human milk. METHODS Milk from lactating mothers was divided into 1 aliquot that was defatted via centrifugation with the full-fat aliquot as control. Macronutrient content was analyzed with mid-infrared spectroscopy. Flow cytometry was used to measure immune cell populations. Lactoferrin, lysozyme, immunoglobulin (Ig)A, and IgG values were determined using enzyme-linked immunosorbent assay. The antibacterial properties were determined by inoculating paired full-fat and defatted milk samples with Escherichia coli or Streptococcus pneumoniae bacteria and performing colony counts. RESULTS Compared with full-fat milk, defatted milk demonstrated decreased total energy and fat and increased carbohydrate concentrations. Defatted milk demonstrated a significant decrease in all immune cell populations. There was no difference in IgA, IgG, lysozyme, or lactoferrin concentrations. Both aliquots demonstrated equivalent growth inhibition of E. coli and S. pneumoniae. CONCLUSIONS Unexpectedly, defatted human milk contained significantly less leukocytes than full-fat milk. IgA, IgG, lysozyme, and lactoferrin concentrations were preserved. The ability of defatted milk to inhibit bacterial growth was unaffected, suggesting that the antibacterial benefits of human milk remain after the defatting process. Further investigation regarding the clinical effect of leukocyte loss in defatted milk is warranted.
Collapse
Affiliation(s)
- Brittany Anne Jackson
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Brigid Ellen Gregg
- Division of Endocrinology, Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Sara Denise Tutor
- Patient Food and Nutrition Services, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jennifer Rachelle Bermick
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Kate Peterson Stanley
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
28
|
Sakwinska O, Bosco N. Host Microbe Interactions in the Lactating Mammary Gland. Front Microbiol 2019; 10:1863. [PMID: 31456777 PMCID: PMC6701204 DOI: 10.3389/fmicb.2019.01863] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/29/2019] [Indexed: 12/13/2022] Open
Abstract
The bacteria present in human milk constitute the human milk microbiome (hMM). Both the older culture-based work and the more recent studies using molecular detection of bacterial DNA have reached similar conclusions: the hMM mostly consists of commensal staphylococci such as Staphylococcus epidermidis, and streptococci. The prevalence of other bacterial groups such lactobacilli varies widely, while the abundance and prevalence of bifidobacteria is generally low. Recently, the hMM became accepted as a part of a physiologically normal state with suggested potential health benefits. Most research on the hMM has focused on its composition and potential effect on the breastfed infant. A major role as a microbiome inoculum for the infant gut has been proposed, but remains to be clearly demonstrated. Herein, we also discuss the emerging connection between the hMM and mammary gland physiology and lactation. Similarities between the mammary gland and mucosal interfaces are considerable, and in particular mucosal-like immune attributes of mammary gland. The potential role of hMM-host interactions in the mammary gland in maternal health is explored with a primary focus on lactational mastitis.
Collapse
Affiliation(s)
- Olga Sakwinska
- Nestlé Research, Nestlé Institute of Health Sciences, Lausanne, Switzerland
| | - Nabil Bosco
- Nestlé Research, Nestlé Institute of Health Sciences, Lausanne, Switzerland
- Nestlé Research Singapore Hub, Singapore, Singapore
| |
Collapse
|
29
|
Nguyen DN, Currie AJ, Ren S, Bering SB, Sangild PT. Heat treatment and irradiation reduce anti-bacterial and immune-modulatory properties of bovine colostrum. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.04.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
30
|
Moreno-Fernandez J, Sánchez-Martínez B, Serrano-López L, Martín-Álvarez E, Diaz-Castro J, Peña-Caballero M, Martín-Peregrina F, Alonso-Moya M, Maldonado-Lozano J, Ochoa JJ, Hurtado-Suazo JA. Enhancement of immune response mediated by oropharyngeal colostrum administration in preterm neonates. Pediatr Allergy Immunol 2019; 30:234-241. [PMID: 30444546 DOI: 10.1111/pai.13008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/29/2018] [Accepted: 10/30/2018] [Indexed: 02/03/2023]
Abstract
BACKGROUND The immune system of preterm infants is immature, being a significant cause of morbidity and mortality, particularly in the preterm infant. Oropharyngeal colostrum administration could be an immunomodulatory aid. Our aim was to evaluate the effect of oropharyngeal colostrum on the serum levels of immunoglobulins, lactoferrin, and resistin during the first month of life and to track the clinical outcome of the neonates. METHODS One hundred preterm neonates born at <32 weeks of gestation and/or weighing < 1500 g and assisted in the Neonatal Intensive Care Unit were enrolled and divided into two groups: colostrum (n = 48) and control (n = 52). The subjects assigned to the colostrum group received 0.2 mL of colostrum (oropharyngeal route) every 4 hours for the first 15 days of life, and if mothers have inability to breastfeed, they were included in the control group (no oropharyngeal colostrum). Serum concentrations of IgA, IgM, and IgG1, lactoferrin, and resistin were assessed in both groups at 1, 3, 15, and 30 days of life. Clinical data during hospitalization were collected. RESULTS IgA and IgM increased in preterm neonates who were administered colostrum for 15 and 30 days. Lactoferrin increased after 30 days, and resistin increased after 15 days of supplying oropharyngeal colostrum. The colostrum group underwent full enteral nutrition before, and no differences were observed in the common neonatal morbidities. CONCLUSION Oropharyngeal colostrum administration is safe in preterm neonates and improves their immunologic profile, showing a potential role as an immunomodulatory agent.
Collapse
Affiliation(s)
- Jorge Moreno-Fernandez
- Department of Physiology, University of Granada, Granada, Spain.,Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, Granada, Spain
| | - Belén Sánchez-Martínez
- Unit of Neonatology, Pediatric Service, Hospital Universitario Materno-Infantil Virgen de las Nieves, Granada, Spain
| | - Laura Serrano-López
- Unit of Neonatology, Pediatric Service, Hospital Universitario Materno-Infantil Virgen de las Nieves, Granada, Spain
| | - Estefanía Martín-Álvarez
- Unit of Neonatology, Pediatric Service, Hospital Universitario Materno-Infantil Virgen de las Nieves, Granada, Spain
| | - Javier Diaz-Castro
- Department of Physiology, University of Granada, Granada, Spain.,Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, Granada, Spain
| | - Manuela Peña-Caballero
- Unit of Neonatology, Pediatric Service, Hospital Universitario Materno-Infantil Virgen de las Nieves, Granada, Spain
| | - Francisca Martín-Peregrina
- Unit of Neonatology, Pediatric Service, Hospital Universitario Materno-Infantil Virgen de las Nieves, Granada, Spain
| | - Mercedes Alonso-Moya
- Unit of Neonatology, Pediatric Service, Hospital Universitario Materno-Infantil Virgen de las Nieves, Granada, Spain
| | - José Maldonado-Lozano
- Pediatrics Department, Virgen de las Nieves University Hospital, University of Granada, Granada, Spain.,Institute of Biosanitary Research of Granada, Maternal and Child Health Network, Carlos III Institute, Madrid, Spain
| | - Julio J Ochoa
- Department of Physiology, University of Granada, Granada, Spain.,Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, Granada, Spain
| | - Jose A Hurtado-Suazo
- Unit of Neonatology, Pediatric Service, Hospital Universitario Materno-Infantil Virgen de las Nieves, Granada, Spain
| |
Collapse
|
31
|
Elwakiel M, Boeren S, Hageman JA, Szeto IM, Schols HA, Hettinga KA. Variability of Serum Proteins in Chinese and Dutch Human Milk during Lactation. Nutrients 2019; 11:E499. [PMID: 30818777 PMCID: PMC6471199 DOI: 10.3390/nu11030499] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/21/2019] [Accepted: 02/24/2019] [Indexed: 01/22/2023] Open
Abstract
To better understand the variability of the type and level of serum proteins in human milk, the milk serum proteome of Chinese mothers during lactation was investigated using proteomic techniques and compared to the milk serum proteome of Dutch mothers. This showed that total milk serum protein concentrations in Chinese human milk decreased over a 20-week lactation period, although with variation between mothers in the rate of decrease. Variation was also found in the composition of serum proteins in both colostrum and mature milk, although immune-active proteins, enzymes, and transport proteins were the most abundant for all mothers. These three protein groups account for many of the 15 most abundant proteins, with these 15 proteins covering more than 95% of the total protein concentrations, in both the Chinese and Dutch milk serum proteome. The Dutch and Chinese milk serum proteome were also compared based on 166 common milk serum proteins, which showed that 22% of the 166 serum proteins differed in level. These differences were observed mainly in colostrum and concern several highly abundant proteins. This study also showed that protease inhibitors, which are highly correlated to immune-active proteins, are present in variable amounts in human milk and could be relevant during digestion.
Collapse
Affiliation(s)
- Mohèb Elwakiel
- Food Quality and Design Group, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands.
- Laboratory of Food Chemistry, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands.
| | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands.
| | - Jos A Hageman
- Biometris-Applied Statistics, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands.
| | - Ignatius M Szeto
- Inner Mongolia Yili Industrial Group Co., Ltd., Jinshan Road 8, Hohhot 010110, China.
| | - Henk A Schols
- Laboratory of Food Chemistry, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands.
| | - Kasper A Hettinga
- Food Quality and Design Group, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands.
| |
Collapse
|
32
|
Alinaghi M, Jiang PP, Brunse A, Sangild PT, Bertram HC. Rapid Cerebral Metabolic Shift during Neonatal Sepsis Is Attenuated by Enteral Colostrum Supplementation in Preterm Pigs. Metabolites 2019; 9:metabo9010013. [PMID: 30641988 PMCID: PMC6359096 DOI: 10.3390/metabo9010013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/04/2019] [Accepted: 01/07/2019] [Indexed: 12/18/2022] Open
Abstract
Sepsis, the clinical manifestation of serious infection, may disturb normal brain development, especially in preterm infants with an immature brain. We hypothesized that neonatal sepsis induces systemic metabolic alterations that rapidly affect metabolic signatures in immature brain and cerebrospinal fluid (CSF). Cesarean-delivered preterm pigs systemically received 109 CFU/kg Staphylococcus epidermidis (SE) and were provided total parenteral nutrition (n = 9) or enteral supplementation with bovine colostrum (n = 10) and compared with uninfected pigs receiving parenteral nutrition (n = 7). Plasma, CSF, and brain tissue samples were collected after 24 h and analyzed by 1H NMR-based metabolomics. Both plasma and CSF metabolomes revealed SE-induced changes in metabolite levels that reflected a modified energy metabolism. Hence, increased plasma lactate, alanine, and succinate levels, as well as CSF lactate levels, were observed during SE infection (all p < 0.05, ANOVA analysis). Myo-inositol, a glucose derivative known for beneficial effects on lung maturation in preterm infants, was also increased in plasma and CSF following SE infection. Enteral colostrum supplementation attenuated the lactate accumulation in blood and CSF. Bloodstream infection in preterm newborns was found to induce a rapid metabolic shift in both plasma and CSF, which was modulated by colostrum feeding.
Collapse
Affiliation(s)
- Masoumeh Alinaghi
- Department of Food Science, Aarhus University, Kirstinebjergvej 10, 5792 Aarslev, Denmark.
| | - Ping-Ping Jiang
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark.
- School of Public Health, Sun Yat-sen University, 510220 Guangzhou, China.
| | - Anders Brunse
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark.
| | - Per Torp Sangild
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark.
| | | |
Collapse
|
33
|
NICU human milk dose and health care use after NICU discharge in very low birth weight infants. J Perinatol 2019; 39:120-128. [PMID: 30341399 PMCID: PMC6298834 DOI: 10.1038/s41372-018-0246-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 09/14/2018] [Accepted: 09/18/2018] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To determine the association between human milk (HM) dose and health care utilization at one and 2 years of life in very low birth weight (birth weight < 1500 g; VLBW) infants. STUDY DESIGN This study included 345 VLBW infants enrolled in a prospective observational cohort study (2008-2012) who completed a neonatal high-risk follow-up clinic visit. Subsequent health care utilization included hospitalizations, emergency department visits, pediatric subspecialists, and specialized therapies. RESULTS Each 10 mL/kg/day increase in HM in the first 14 days of life was associated with 0.26 fewer hospitalizations (p = 0.04) at 1 year and 0.21 fewer pediatric subspecialist types (p = 0.04) and 0.20 fewer specialized therapy types (p = 0.04) at 2 years. CONCLUSION HM dose in early life for VLBW infants was an independent predictor of the number of hospitalizations at 1 year and types of pediatric subspecialists and specialized therapies at 2 years of life.
Collapse
|
34
|
van den Elsen LWJ, Garssen J, Burcelin R, Verhasselt V. Shaping the Gut Microbiota by Breastfeeding: The Gateway to Allergy Prevention? Front Pediatr 2019; 7:47. [PMID: 30873394 PMCID: PMC6400986 DOI: 10.3389/fped.2019.00047] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/04/2019] [Indexed: 12/23/2022] Open
Abstract
Evidence is accumulating that demonstrates the importance of the gut microbiota in health and diseases such as allergy. Recent studies emphasize the importance of the "window of opportunity" in early life, during which interventions altering the gut microbiota induce long-term effects. The neonate's gut microbiota composition and metabolism could therefore play an essential role in allergic disease risk. Breastfeeding shapes the gut microbiota in early life, both directly by exposure of the neonate to the milk microbiota and indirectly, via maternal milk factors that affect bacterial growth and metabolism such as human milk oligosaccharides, secretory IgA, and anti-microbial factors. The potential of breastmilk to modulate the offspring's early gut microbiota is a promising tool for allergy prevention. Here, we will review the existing evidence demonstrating the impact of breastfeeding on shaping the neonate's gut microbiota and highlight the potential of this strategy for allergy prevention.
Collapse
Affiliation(s)
| | - Johan Garssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Remy Burcelin
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
| | - Valerie Verhasselt
- School of Molecular Sciences, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
35
|
Effects of lactoferrin on neonatal pathogens and Bifidobacterium breve in human breast milk. PLoS One 2018; 13:e0201819. [PMID: 30133470 PMCID: PMC6104981 DOI: 10.1371/journal.pone.0201819] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 07/22/2018] [Indexed: 01/22/2023] Open
Abstract
Supplementation with probiotics in preterm infants reduces necrotizing enterocolitis and sepsis. Bovine lactoferrin is a promising supplement that may further reduce disease burden, but its effects on probiotic bacteria in human breast milk has not been evaluated. We aimed to characterise the antimicrobial activity of bovine and human lactoferrin in human breast milk against probiotics and typical neonatal sepsis pathogens. Lactoferrin levels were determined by enzyme linked immunosorbent assay in fresh and pasteurised human breast milk. The neonatal pathogens Staphylococcus epidermidis and Escherichia coli, and the probiotic Bifidobacterium breve strain M-16V were cultured in human breast milk or infant formula in the presence or absence of clinically relevant doses of bovine or human lactoferrin. Standard microbiological methods were used to determine the effects of lactoferrin on bacterial growth. Unpasteurised human breast milk contained significantly higher lactoferrin levels and resulted in superior inhibition of pathogenic bacterial growth compared to infant formula and pasteurised human breast milk. Human lactoferrin was significantly more effective at inhibiting bacterial growth, when compared to bovine lactoferrin. Supplementation with human lactoferrin or high dose bovine lactoferrin inhibited growth of the probiotic strain B. breve M-16V in pasteurised human breast milk. Although unpasteurised human breast milk and human lactoferrin had the greatest antimicrobial activity against all bacterial species tested, higher doses of bovine lactoferrin also showed activity against B. breve and. S. epidermidis. This study suggests that simultaneous administration of lactoferrins and probiotics may affect colonisation with probiotic bacteria, warranting further investigations.
Collapse
|
36
|
Scheid A, Li N, Jeffers C, Borriello F, Joshi S, Ozonoff A, Pettengill M, Levy O. Antimicrobial peptide LL-37 and recombinant human mannose-binding lectin express distinct age- and pathogen-specific antimicrobial activity in human newborn cord blood in vitro. F1000Res 2018; 7:616. [PMID: 30271580 PMCID: PMC6143923 DOI: 10.12688/f1000research.14736.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/10/2018] [Indexed: 01/07/2023] Open
Abstract
Background: There is a need to prevent and treat infection in newborns. One approach is administration of antimicrobial proteins and peptides (APPs) such as LL-37, a membrane-active cathelicidin antimicrobial peptide, and mannose-binding lectin (MBL), a pattern-recognition protein that binds to microbial surface polysaccharides resulting in opsonization and complement activation. Low plasma/serum levels of LL-37 and of MBL have been correlated with infection and exogenous administration of these agents may enhance host defense. Methods: The antimicrobial activity of LL-37 (15 µg/ml) or rMBL (0.5, 2 and 10 µg/ml) was tested in hirudin-anticoagulated preterm and term human cord blood (N = 12-14) against Staphylococcus aureus (SA) USA 300 (2x10 4 CFU/ml), Staphylococcus epidermis (SE) 1457 (2x10 4 CFU/ml) and Candida albicans (CA) SC5314 (1x10 4 CFU/ml). After incubation (1, 45, or 180 min), CFUs were enumerated by plating blood onto agar plates. Supernatants were collected for measurement of MBL via ELISA. Results: Preterm cord blood demonstrated impaired endogenous killing capacity against SA and SE compared to term blood. Addition of LL-37 strongly enhanced antimicrobial/antifungal activity vs SA, SE and CA in term blood and SE and CA in preterm blood. By contrast, rMBL showed modest fungistatic activity vs CA in a sub-analysis of term newborns with high basal MBL levels. Baseline MBL levels varied within preterm and term cohorts with no correlation to gestational age. In summary, exogenous LL-37 demonstrated significant antimicrobial activity against SA, SE and CA in term and SE and CA in preterm human blood tested in vitro. rMBL demonstrated modest antifungal activity in term cord blood of individuals with high baseline MBL levels. Conclusions: To the extent that our in vitro results predict the effects of APPs in vivo, development of APPs for prevention and treatment of infection should take into account host age as well as the target pathogen.
Collapse
Affiliation(s)
- Annette Scheid
- Department of Pediatric Newborn Medicine, Brigham and Women's University Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Ning Li
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Carleen Jeffers
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Francesco Borriello
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
- WAO Center of Excellence, Naples, Italy
| | - Sweta Joshi
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Al Ozonoff
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Center for Patient Safety and Quality Research, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Matthew Pettengill
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT & Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
37
|
Pirr S, Richter M, Fehlhaber B, Pagel J, Härtel C, Roth J, Vogl T, Viemann D. High Amounts of S100-Alarmins Confer Antimicrobial Activity on Human Breast Milk Targeting Pathogens Relevant in Neonatal Sepsis. Front Immunol 2017; 8:1822. [PMID: 29326708 PMCID: PMC5733341 DOI: 10.3389/fimmu.2017.01822] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 12/04/2017] [Indexed: 12/15/2022] Open
Abstract
Sepsis is a leading cause of perinatal mortality worldwide. Breast milk (BM) feeding is protective against neonatal sepsis, but the molecular mechanisms remain unexplained. Despite various supplementations with potential bioactive components from BM formula feeding cannot protect from sepsis. S100-alarmins are important immunoregulators in newborns preventing excessive inflammation. At high concentrations, the S100A8/A9 protein complex also has antimicrobial properties due to its metal ion chelation capacity. To assess whether BM contains S100-alarmins that might mediate the sepsis-protective effect of BM 97 human BM samples stratified for gestational age, mode of delivery and sampling after birth were collected and analyzed. S100A8/A9 levels were massively elevated after birth (p < 0.0005). They slowly decreased during the first month of life, then reaching levels comparable to normal values in adult serum. The concentration of S100A8/A9 in BM was significantly higher after term compared with preterm birth (extremely preterm, p < 0.005; moderate preterm, p < 0.05) and after vaginal delivery compared with cesarean section (p < 0.0005). In newborn s100a9−/− mice, enterally supplied S100-alarmins could be retrieved systemically in the plasma. To explore the antimicrobial activity against common causal pathogens of neonatal sepsis, purified S100-alarmins and unmodified as well as S100A8/A9-depleted BM were used in growth inhibition tests. The high amount of S100A8/A9 proved to be an important mediator of the antimicrobial activity of BM, especially inhibiting the growth of manganese (Mn) sensitive bacteria such as Staphylococcus aureus (p < 0.00005) and group B streptococci (p < 0.005). Depletion of S100A8/A9 significantly reduced this effect (p < 0.05, respectively). The growth of Escherichia coli was also inhibited by BM (p < 0.00005) as well as by S100A8/A9 in culture assays (p < 0.05). But its growth in BM remained unaffected by the removal of S100A8/A9 and was neither dependent on Mn suggesting that the antimicrobial effects of S100A8/A9 in BM are primarily mediated by its Mn chelating capacity. In summary, the enteral supply of bioavailable, antimicrobially active amounts of S100-alarmins might be a promising option to protect newborns at high risk from infections and sepsis.
Collapse
Affiliation(s)
- Sabine Pirr
- Department of Pediatric Pneumology, Allergy and Neonatology, Hannover Medical School, Hannover, Germany
| | - Manuela Richter
- Department of Pediatric Pneumology, Allergy and Neonatology, Hannover Medical School, Hannover, Germany.,Children's Hospital "Auf der Bult", Hannover, Germany
| | - Beate Fehlhaber
- Department of Pediatric Pneumology, Allergy and Neonatology, Hannover Medical School, Hannover, Germany
| | - Julia Pagel
- Department of Pediatrics, University of Lübeck, Lübeck, Germany
| | | | - Johannes Roth
- Institute of Immunology, University of Münster, Münster, Germany
| | - Thomas Vogl
- Institute of Immunology, University of Münster, Münster, Germany
| | - Dorothee Viemann
- Department of Pediatric Pneumology, Allergy and Neonatology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
38
|
Fu Y, Ji C, Chen X, Cui X, Wang X, Feng J, Li Y, Qin R, Guo X. Investigation into the antimicrobial action and mechanism of a novel endogenous peptide β-casein 197 from human milk. AMB Express 2017; 7:119. [PMID: 28591979 PMCID: PMC5461228 DOI: 10.1186/s13568-017-0409-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 05/23/2017] [Indexed: 12/01/2022] Open
Abstract
A novel endogenous peptide cleaved from 197–213 AA of β-casein, named β-casein 197, was identified by tandem mass spectrometry. β-casein 197 constituted a significant proportion of the peptide content in preterm milk. This study investigated the antibacterial effects and mechanisms against common pathogenic bacteria. Six bacterial strains were selected for this study: Escherichia coli, Staphylococcus aureus, Yersinia enterocolitica, Listeria monocytogenes, Klebsiella pneumonia and Bacillus subtilis. After synthesis, serial twofold dilutions of β-casein 197 were added to select for sensitive bacteria. The disk diffusion method and analysis of bacterial staining were used to identify antibacterial effect, while DNA-binding, scanning electron microscopy and transmission electron microscopy were used to explore antimicrobial mechanisms. Disk diffusion showed that E. coli, S. aureus and Y. enterocolitica were sensitive to the β-casein 197. In addition, live/dead fluorescent staining also confirmed antibacterial effects. Scanning electron and transmission electron microscopy revealed affected extracellular and intracellular structure for three species of bacteria, while a DNA-binding assay showed that the antimicrobial activity did not occur through DNA binding. This study suggests that β-casein 197 has antimicrobial activity against common pathogenic bacteria in newborns with infection. The peptide induced membrane permeabilization but did not bind to genomic DNA. Based on our findings, β-casein 197 has potential clinical value for preventing infections of premature infants.
Collapse
|
39
|
Strunk T, Hibbert J, Doherty D, Granland C, Trend S, Simmer K, Burgner D, Patole S, Currie A. Probiotics and antimicrobial protein and peptide levels in preterm infants. Acta Paediatr 2017; 106:1747-1753. [PMID: 28294428 DOI: 10.1111/apa.13826] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/22/2017] [Accepted: 03/07/2017] [Indexed: 12/26/2022]
Abstract
AIM To characterise the secreted and inducible antimicrobial protein and peptide (APP) levels in a prospective cohort of preterm infants (<30 weeks gestational age) with or without Bifidobacterium breve M16V supplementation during the first month of life. METHODS We analysed serial biosamples of infants who did (n = 13) or did not receive (n = 62) B. breve (3 × 109 cfu/day). Peripheral blood was obtained on days 1, 14 and 28, and infant stool prior to commencement of probiotic supplementation and on day 21. Levels of APP (bactericidal/permeability inducing protein (BPI), beta defensins 1 and 2, lactoferrin, human cathelicidin, secretory phospholipase A2) in plasma and stool were determined. Further, we characterised induced APP levels in whole blood cultured with live S. epidermidis or with agonists of Toll-like receptors 2/6 and 4. RESULTS Stool, plasma and stimulated blood APP levels changed significantly during the first month of life. Supplementation with B. breve did not affect basal or stimulated APP levels except for a transient increase in inducible BPI. CONCLUSION Supplementation with B. breve does not appear to act via modulation of systemic or enteric APP expression in preterm infants although small effects cannot be excluded. Further work with other probiotic preparations is warranted.
Collapse
Affiliation(s)
- Tobias Strunk
- Centre for Neonatal Research and Education; School of Paediatrics and Child Health; University of Western Australia; Perth WA Australia
- Neonatal Clinical Care Unit; King Edward Memorial Hospital; Perth WA Australia
| | - Julie Hibbert
- Centre for Neonatal Research and Education; School of Paediatrics and Child Health; University of Western Australia; Perth WA Australia
| | - Dorota Doherty
- School of Women's and Infant's Health; University of Western Australia; Perth WA Australia
| | - Caitlyn Granland
- Centre for Neonatal Research and Education; School of Paediatrics and Child Health; University of Western Australia; Perth WA Australia
| | - Stephanie Trend
- Centre for Neonatal Research and Education; School of Paediatrics and Child Health; University of Western Australia; Perth WA Australia
| | - Karen Simmer
- Centre for Neonatal Research and Education; School of Paediatrics and Child Health; University of Western Australia; Perth WA Australia
- Neonatal Clinical Care Unit; King Edward Memorial Hospital; Perth WA Australia
| | - David Burgner
- Murdoch Childrens Research Institute; Royal Children's Hospital; Parkville VIC Australia
- Department of Paediatrics; University of Melbourne; Melbourne VIC Australia
| | - Sanjay Patole
- Centre for Neonatal Research and Education; School of Paediatrics and Child Health; University of Western Australia; Perth WA Australia
- Neonatal Clinical Care Unit; King Edward Memorial Hospital; Perth WA Australia
| | - Andrew Currie
- Centre for Neonatal Research and Education; School of Paediatrics and Child Health; University of Western Australia; Perth WA Australia
- School of Veterinary and Life Sciences; Murdoch University; Perth WA Australia
| |
Collapse
|
40
|
Hering NA, Luettig J, Krug SM, Wiegand S, Gross G, van Tol EA, Schulzke JD, Rosenthal R. Lactoferrin protects against intestinal inflammation and bacteria-induced barrier dysfunction in vitro. Ann N Y Acad Sci 2017; 1405:177-188. [PMID: 28614589 DOI: 10.1111/nyas.13405] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 05/05/2017] [Accepted: 05/10/2017] [Indexed: 12/15/2022]
Abstract
The iron-binding glycoprotein lactoferrin (LF) is naturally present in human breast milk. Several studies suggest that LF contributes to infant health and development owing to a variety of protective effects, including antimicrobial and anti-inflammatory features. Therefore, we aimed to elucidate its protective properties on intestinal epithelial barrier dysfunction induced by infection or inflammation using the human epithelial cell culture models HT-29/B6 and T84. During barrier perturbation induced by the proinflammatory cytokine tumor necrosis factor α (TNF-α), bovine LF restored tight junction (TJ) morphometry and inhibited TNF-α-induced epithelial apoptosis. This resulted in an attenuation of the TNF-α-induced decrease in transepithelial resistance (TER) and increases in permeability of fluorescein and FITC-dextran (4 kDa) and was as effective as the apoptosis inhibitor Q-VD-Oph. The enteropathogenic bacterium Yersinia enterocolitica is a frequent cause of diarrhea in early childhood. This involves focal changes in TJ protein expression and localization. LF diminished the Y. enterocolitica-induced drop in TER in the present in vitro model, which was paralleled by an inhibition of the Yersinia-induced reduction of claudin-8 expression via c-Jun kinase signaling. In conclusion, LF exerts protective effects against inflammation- or infection-induced barrier dysfunction in human intestinal cell lines, supporting its relevance for healthy infant development.
Collapse
Affiliation(s)
- Nina A Hering
- Department of General, Visceral and Vascular Surgery, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Luettig
- Institute of Clinical Physiology, Department of Gastroenterology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Susanne M Krug
- Institute of Clinical Physiology, Department of Gastroenterology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stephanie Wiegand
- Institute of Clinical Physiology, Department of Gastroenterology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Gabriele Gross
- Mead Johnson Pediatric Nutrition Institute, Nijmegen, the Netherlands
| | - Eric A van Tol
- Mead Johnson Pediatric Nutrition Institute, Nijmegen, the Netherlands
| | - Jörg D Schulzke
- Institute of Clinical Physiology, Department of Gastroenterology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Rita Rosenthal
- Institute of Clinical Physiology, Department of Gastroenterology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
41
|
Cacho NT, Lawrence RM. Innate Immunity and Breast Milk. Front Immunol 2017; 8:584. [PMID: 28611768 PMCID: PMC5447027 DOI: 10.3389/fimmu.2017.00584] [Citation(s) in RCA: 247] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/01/2017] [Indexed: 12/16/2022] Open
Abstract
Human milk is a dynamic source of nutrients and bioactive factors; unique in providing for the human infant's optimal growth and development. The growing infant's immune system has a number of developmental immune deficiencies placing the infant at increased risk of infection. This review focuses on how human milk directly contributes to the infant's innate immunity. Remarkable new findings clarify the multifunctional nature of human milk bioactive components. New research techniques have expanded our understanding of the potential for human milk's effect on the infant that will never be possible with milk formulas. Human milk microbiome directly shapes the infant's intestinal microbiome, while the human milk oligosaccharides drive the growth of these microbes within the gut. New techniques such as genomics, metabolomics, proteomics, and glycomics are being used to describe this symbiotic relationship. An expanded role for antimicrobial proteins/peptides within human milk in innate immune protection is described. The unique milieu of enhanced immune protection with diminished inflammation results from a complex interaction of anti-inflammatory and antioxidative factors provided by human milk to the intestine. New data support the concept of mucosal-associated lymphoid tissue and its contribution to the cellular content of human milk. Human milk stem cells (hMSCs) have recently been discovered. Their direct role in the infant for repair and regeneration is being investigated. The existence of these hMSCs could prove to be an easily harvested source of multilineage stem cells for the study of cancer and tissue regeneration. As the infant's gastrointestinal tract and immune system develop, there is a comparable transition in human milk over time to provide fewer immune factors and more calories and nutrients for growth. Each of these new findings opens the door to future studies of human milk and its effect on the innate immune system and the developing infant.
Collapse
Affiliation(s)
- Nicole Theresa Cacho
- Division of Neonatology, Department of Pediatrics, University of Florida, Gainesville, FL, United States
| | - Robert M Lawrence
- Division of Pediatric Infectious Disease, Department of Pediatrics, University of Florida, Gainesville, FL, United States
| |
Collapse
|
42
|
Zambruni M, Villalobos A, Somasunderam A, Westergaard S, Nigalye M, Turin CG, Zegarra J, Bellomo S, Mercado E, Ochoa TJ, Utay NS. Maternal and pregnancy-related factors affecting human milk cytokines among Peruvian mothers bearing low-birth-weight neonates. J Reprod Immunol 2017; 120:20-26. [PMID: 28399439 PMCID: PMC5494983 DOI: 10.1016/j.jri.2017.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 03/17/2017] [Accepted: 04/03/2017] [Indexed: 12/13/2022]
Abstract
Several cytokines have been detected in human milk but their relative concentrations differ among women and vary over time in the same person. The drivers of such differences have been only partially identified, while the effect of luminal cytokines in the fine-regulation of the intestinal immune system is increasingly appreciated. The aim of this study was to investigate the associations between obstetrical complications and human milk cytokine profiles in a cohort of Peruvian women giving birth to Low Birth Weight (LBW) infants. Colostrum and mature human milk samples were collected from 301 Peruvian women bearing LBW infants. The concentration of twenty-three cytokines was measured using the Luminex platform. Ninety-nine percent of women had at least one identified obstetrical complication leading to intra-uterine growth restriction and/or preterm birth. Median weight at birth was 1,420g; median gestational age 31 weeks. A core of 12 cytokines, mainly involved in innate immunity and epithelial cell integrity, was detectable in most samples. Maternal age, maternal infection, hypertensive disorders, preterm labor, and premature rupture of membranes were associated with specific cytokine profiles both in colostrum and mature human milk. Mothers of Very LBW (VLBW) neonates had significantly higher concentrations of chemokines and growth factor cytokines both in their colostrum and mature milk compared with mothers of larger neonates. Thus, maternal conditions affecting pregnancy duration and in utero growth are also associated with specific human milk cytokine signatures.
Collapse
Affiliation(s)
- Mara Zambruni
- University of Texas Medical Branch (UTMB), Department of Internal Medicine, Division of Infectious Diseases; Galveston, TX, USA
| | - Alex Villalobos
- University of Texas Medical Branch (UTMB), School of Medicine, Galveston, TX, USA
| | - Anoma Somasunderam
- University of Texas Medical Branch (UTMB), Department of Internal Medicine, Division of Infectious Diseases; Galveston, TX, USA
| | - Sarah Westergaard
- University of Texas Medical Branch (UTMB), School of Medicine, Galveston, TX, USA
| | - Maitreyee Nigalye
- University of Texas Medical Branch (UTMB), Department of Internal Medicine, Division of Infectious Diseases; Galveston, TX, USA
| | - Christie G Turin
- Universidad Peruana Cayetano Heredia, Laboratorio de Infectologia Pediatrica, Lima, Peru
| | - Jaime Zegarra
- Universidad Peruana Cayetano Heredia, Department of Pediatrics, Lima, Peru
| | - Sicilia Bellomo
- Universidad Peruana Cayetano Heredia, Department of Pediatrics, Lima, Peru
| | - Erik Mercado
- Universidad Peruana Cayetano Heredia, Laboratorio de Infectologia Pediatrica, Lima, Peru
| | - Theresa J Ochoa
- Universidad Peruana Cayetano Heredia, Department of Pediatrics, Lima, Peru; University of Texas Health Science Center at Houston (UTHSCH), Houston, TX, USA
| | - Netanya S Utay
- University of Texas Medical Branch (UTMB), Department of Internal Medicine, Division of Infectious Diseases; Galveston, TX, USA.
| |
Collapse
|
43
|
Lourenço AG, Komesu MC, Duarte G, Del Ciampo LA, Mussi-Pinhata MM, Yamamoto AY. High Levels of Chemokine C-C Motif Ligand 20 in Human Milk and Its Production by Oral Keratinocytes. Breastfeed Med 2017; 12:116-121. [PMID: 27912038 DOI: 10.1089/bfm.2016.0067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Chemokine C-C motif ligand 20 (CCL20) is implicated in the formation and function of mucosal lymphoid tissues. Although CCL20 is secreted by many normal human tissues, no studies have evaluated the presence of CCL20 in human milk or its production by oral keratinocytes stimulated by human milk. OBJECTIVE To evaluate the presence of CCL20 in breast milk and verify CCL20 secretion in vitro by oral keratinocytes stimulated with human and bovine milk, as well as its possible association with breast milk lactoferrin levels. MATERIALS AND METHODS The levels of CCL20 and lactoferrin were measured by enzyme-linked immunosorbent assay in human milk at three different stages of maturation from 74 healthy breastfeeding mothers. In vitro, oral keratinocytes were stimulated with human and bovine milk, and CCL20 was measured in their supernatant. RESULTS High concentrations of CCL20 were detected in the human breast milk samples obtained during the first week (1,777.07 pg/mL) and second week postpartum (1,523.44 pg/mL), with a significantly low concentration in samples at 3-6 weeks postpartum (238.42 pg/mL; p < 0.0001). Human breast milk at different weeks postpartum stimulated higher CCL20 secretion by oral keratinocytes compared with bovine milk (p < 0.05). Such stimulation had no association with breast milk lactoferrin concentration. CONCLUSION CCl20 is present at high levels in human milk, predominantly in the first and second week postpartum, but at significantly lower levels at 3-6 weeks postpartum. Human milk is capable of stimulating CCL20 secretion by oral keratinocytes, and this induction had no association with breast milk lactoferrin concentration.
Collapse
Affiliation(s)
- Alan G Lourenço
- 1 Department of Pediatrics, Ribeirão Preto School of Medicine, University of São Paulo , São Paulo, Brazil
| | - Marilena C Komesu
- 2 Department of Morphology, Physiology and Basic Pathology, Ribeirão Preto School of Dentistry, University of São Paulo , São Paulo, Brazil
| | - Geraldo Duarte
- 3 Department of Gynecology and Obstetrics, Ribeirão Preto School of Medicine, University of São Paulo , São Paulo, Brazil
| | - Luiz A Del Ciampo
- 1 Department of Pediatrics, Ribeirão Preto School of Medicine, University of São Paulo , São Paulo, Brazil
| | - Marisa M Mussi-Pinhata
- 1 Department of Pediatrics, Ribeirão Preto School of Medicine, University of São Paulo , São Paulo, Brazil
| | - Aparecida Y Yamamoto
- 1 Department of Pediatrics, Ribeirão Preto School of Medicine, University of São Paulo , São Paulo, Brazil
| |
Collapse
|
44
|
Zhang F, Cui X, Fu Y, Zhang J, Zhou Y, Sun Y, Wang X, Li Y, Liu Q, Chen T. Antimicrobial activity and mechanism of the human milk-sourced peptide Casein201. Biochem Biophys Res Commun 2017; 485:698-704. [PMID: 28242197 DOI: 10.1016/j.bbrc.2017.02.108] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 02/21/2017] [Indexed: 01/30/2023]
Abstract
INTRODUCTION Casein201 is one of the human milk sourced peptides that differed significantly in preterm and full-term mothers. This study is designed to demonstrate the biological characteristics, antibacterial activity and mechanisms of Casein201 against common pathogens in neonatal infection. METHODOLOGY The analysis of biological characteristics was done by bioinformatics. Disk diffusion method and flow cytometry were used to detect the antimicrobial activity of Casein201. Killing kinetics of Casein201 was measured using microplate reader. The antimicrobial mechanism of Casein201 was studied by electron microscopy and electrophoresis. RESULTS Bioinformatics analysis indicates that Casein201 derived from β-casein and showed significant sequence overlap. Antibacterial assays showed Casein201 inhibited the growth of S taphylococcus aureus and Y ersinia enterocolitica. Ultrastructural analyses revealed that the antibacterial activity of Casein201 is through cytoplasmic structures disintegration and bacterial cell envelope alterations but not combination with DNA. CONCLUSION We conclude the antimicrobial activity and mechanism of Casein201. Our data demonstrate that Casein201 has potential therapeutic value for the prevention and treatment of pathogens in neonatal infection.
Collapse
Affiliation(s)
- Fan Zhang
- Nanjing Maternal and Child Health Medical Institute, Nanjing Maternal and Child Health Hospital, Obestetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, China; Department of Endocrinology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xianwei Cui
- Nanjing Maternal and Child Health Medical Institute, Nanjing Maternal and Child Health Hospital, Obestetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yanrong Fu
- Nanjing Maternal and Child Health Medical Institute, Nanjing Maternal and Child Health Hospital, Obestetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Jun Zhang
- Nanjing Maternal and Child Health Medical Institute, Nanjing Maternal and Child Health Hospital, Obestetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yahui Zhou
- Nanjing Maternal and Child Health Medical Institute, Nanjing Maternal and Child Health Hospital, Obestetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yazhou Sun
- Nanjing Maternal and Child Health Medical Institute, Nanjing Maternal and Child Health Hospital, Obestetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Xing Wang
- Nanjing Maternal and Child Health Medical Institute, Nanjing Maternal and Child Health Hospital, Obestetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yun Li
- Nanjing Maternal and Child Health Medical Institute, Nanjing Maternal and Child Health Hospital, Obestetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Qianqi Liu
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Nanjing, China.
| | - Ting Chen
- Nanjing Maternal and Child Health Medical Institute, Nanjing Maternal and Child Health Hospital, Obestetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, China.
| |
Collapse
|
45
|
Kothary V, Doster RS, Rogers LM, Kirk LA, Boyd KL, Romano-Keeler J, Haley KP, Manning SD, Aronoff DM, Gaddy JA. Group B Streptococcus Induces Neutrophil Recruitment to Gestational Tissues and Elaboration of Extracellular Traps and Nutritional Immunity. Front Cell Infect Microbiol 2017; 7:19. [PMID: 28217556 PMCID: PMC5289994 DOI: 10.3389/fcimb.2017.00019] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 01/16/2017] [Indexed: 12/14/2022] Open
Abstract
Streptococcus agalactiae, or Group B Streptococcus (GBS), is a gram-positive bacterial pathogen associated with infection during pregnancy and is a major cause of morbidity and mortality in neonates. Infection of the extraplacental membranes surrounding the developing fetus, a condition known as chorioamnionitis, is characterized histopathologically by profound infiltration of polymorphonuclear cells (PMNs, neutrophils) and greatly increases the risk for preterm labor, stillbirth, or neonatal GBS infection. The advent of animal models of chorioamnionitis provides a powerful tool to study host-pathogen relationships in vivo and ex vivo. The purpose of this study was to evaluate the innate immune response elicited by GBS and evaluate how antimicrobial strategies elaborated by these innate immune cells affect bacteria. Our work using a mouse model of GBS ascending vaginal infection during pregnancy reveals that clinically isolated GBS has the capacity to invade reproductive tissues and elicit host immune responses including infiltration of PMNs within the choriodecidua and placenta during infection, mirroring the human condition. Upon interacting with GBS, murine neutrophils elaborate DNA-containing extracellular traps, which immobilize GBS and are studded with antimicrobial molecules including lactoferrin. Exposure of GBS to holo- or apo-forms of lactoferrin reveals that the iron-sequestration activity of lactoferrin represses GBS growth and viability in a dose-dependent manner. Together, these data indicate that the mouse model of ascending infection is a useful tool to recapitulate human models of GBS infection during pregnancy. Furthermore, this work reveals that neutrophil extracellular traps ensnare GBS and repress bacterial growth via deposition of antimicrobial molecules, which drive nutritional immunity via metal sequestration strategies.
Collapse
Affiliation(s)
- Vishesh Kothary
- Department of Medicine, Vanderbilt University School of Medicine Nashville, TN, USA
| | - Ryan S Doster
- Department of Medicine, Vanderbilt University Medical Center Nashville, TN, USA
| | - Lisa M Rogers
- Department of Medicine, Vanderbilt University Medical Center Nashville, TN, USA
| | - Leslie A Kirk
- Department of Medicine, Vanderbilt University Medical Center Nashville, TN, USA
| | - Kelli L Boyd
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Joann Romano-Keeler
- Department of Pediatrics, Vanderbilt University Medical Center Nashville, TN, USA
| | - Kathryn P Haley
- Department of Medicine, Vanderbilt University Medical CenterNashville, TN, USA; Department of Biomedical Sciences, Grand Valley State UniversityGrand Rapids, MI, USA
| | - Shannon D Manning
- Department of Microbiology and Molecular Genetics, Michigan State University East Lansing, MI, USA
| | - David M Aronoff
- Department of Medicine, Vanderbilt University School of Medicine Nashville, TN, USA
| | - Jennifer A Gaddy
- Department of Medicine, Vanderbilt University Medical CenterNashville, TN, USA; Department of Veterans Affairs, Tennessee Valley Healthcare SystemsNashville, TN, USA
| |
Collapse
|
46
|
Haj Ebrahim Tehrani F, Moradi M, Ghorbani N. Bacterial Etiology and Antibiotic Resistance Patterns in Neonatal Sepsis in Tehran during 2006-2014. IRANIAN JOURNAL OF PATHOLOGY 2017; 12:356-361. [PMID: 29563931 PMCID: PMC5844680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 01/25/2016] [Indexed: 11/30/2022]
Abstract
BACKGROUND & OBJECTIVE Neonatal sepsis is one of the leading causes of mortality in neonatology wards. The aim of this study was to assess sepsis pathogens and antibacterial resistance patterns in a teaching hospital during seven years in Tehran, Iran. METHODS In this retrospective study, all neonates suspected to sepsis and fulfilling the sepsis criteria admitted to NICU ward of Mustafa Khomeini Hospital, Tehran, Iran during 2007 to 2014 were included. Demographic information, blood test results, blood culture results of neonates and antibiogram findings were extracted from their documents. Data was analyzed using SPSS 15. RESULTS Ninety neonates with positive culture test were included. Fifty-three were male (58.9%). Thirty neonates were delivered vaginally (33.3%) and 60 caesarean section (66.7%). Most bacterial growths in culture were Staphylococcus aureus and E. coli. The rates of resistance for antibiotics like ceftriaxone, cefotaxim and gentamycin were 5%, 30% and 15%, correspondingly. There were 15 cases (16.7%) with resistance to imipenem. CONCLUSION Antibacterial resistance patterns vary in different parts of the world and even within a country, therefore assessing resistance patterns in a region is of great importance for proper management and treatment. Our findings might help physicians for proper selection of antibiotics for treatment of neonatal sepsis.
Collapse
Affiliation(s)
| | - Mohammad Moradi
- Students Research Committee of Shahed Medical Faculty, Shahed University, Tehran, Iran,Corresponding information: Dr. Mohammad Moradi, Students Research Committee of Shahed Medical Faculty, Shahed University, Tehran, Iran, E-mail:
| | | |
Collapse
|
47
|
Decembrino L, DeAmici M, De Silvestri A, Manzoni P, Paolillo P, Stronati M. Plasma lactoferrin levels in newborn preterm infants with sepsis. J Matern Fetal Neonatal Med 2016; 30:2890-2893. [DOI: 10.1080/14767058.2016.1266479] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Lidia Decembrino
- Fondazione IRCCS Policlinico San Matteo, Neonatal Intesive Care Unit, Pavia, Italy
| | - Mara DeAmici
- Fondazione IRCCS Policlinico San Matteo, Pediatric Clinic, IRCCS San Matteo Foundation, Pavia, Italy
| | | | - Paolo Manzoni
- Neonatology and Neonatal Intensive Care Unit, S. Anna Hospital, Torino, Italy
| | | | - Mauro Stronati
- Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
48
|
Battersby AJ, Khara J, Wright VJ, Levy O, Kampmann B. Antimicrobial Proteins and Peptides in Early Life: Ontogeny and Translational Opportunities. Front Immunol 2016; 7:309. [PMID: 27588020 PMCID: PMC4989132 DOI: 10.3389/fimmu.2016.00309] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 07/29/2016] [Indexed: 12/18/2022] Open
Abstract
While developing adaptive immune responses, young infants are especially vulnerable to serious infections, including sepsis, meningitis, and pneumonia. Antimicrobial proteins and peptides (APPs) are key effectors that function as broad-spectrum anti-infectives. This review seeks to summarize the clinically relevant functional qualities of APPs and the increasing clinical trial evidence for their use to combat serious infections in infancy. Levels of APPs are relatively low in early life, especially in infants born preterm or with low birth weight (LBW). There are several rationales for the potential clinical utility of APPs in the prevention and treatment of infections in infants: (a) APPs may be most helpful in those with reduced levels; (b) during sepsis microbial products signal via pattern recognition receptors causing potentially harmful inflammation that APPs may counteract; and (c) in the era of antibiotic resistance, development of new anti-infective strategies is essential. Evidence supports the potential clinical utility of exogenous APPs to reduce infection-related morbidity in infancy. Further studies should characterize the ontogeny of antimicrobial activity in mucosal and systemic compartments, and examine the efficacy of exogenous-APP formulations to inform translational development of APPs for infant groups.
Collapse
Affiliation(s)
- Anna J Battersby
- Academic Paediatrics, Imperial College London, London, UK; Medical Research Council (MRC) Unit, Vaccines and Immunity Theme, Fajara, Gambia
| | - Jasmeet Khara
- Academic Paediatrics, Imperial College London, London, UK; Department of Pharmacy, National University of Singapore, Singapore
| | | | - Ofer Levy
- Precision Vaccines Program, Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Beate Kampmann
- Academic Paediatrics, Imperial College London, London, UK; Medical Research Council (MRC) Unit, Vaccines and Immunity Theme, Fajara, Gambia
| |
Collapse
|
49
|
Addis MF, Tedde V, Puggioni GMG, Pisanu S, Casula A, Locatelli C, Rota N, Bronzo V, Moroni P, Uzzau S. Evaluation of milk cathelicidin for detection of bovine mastitis. J Dairy Sci 2016; 99:8250-8258. [PMID: 27522416 DOI: 10.3168/jds.2016-11407] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 06/28/2016] [Indexed: 11/19/2022]
Abstract
Mastitis due to intramammary infection is one of the most economically relevant diseases in dairy cows, causing reductions in milk quality and quantity. Currently, mastitis monitoring is based on somatic cell count (SCC) and bacteriologic culture (BC) of milk. Nevertheless, inflammation-specific protein markers might provide more sensitive and reliable assays, enabling immunoassay-based screening strategies. Cathelicidin is an inflammatory protein released in milk that has recently demonstrated fair reliability and diagnostic potential for ewe mastitis. To assess its performance in cows, 531 quarter milk samples from 2 herds were tested using cathelicidin ELISA, SCC, and BC. We found that 29.0% of samples were positive for cathelicidin, 18.8% had SCC >200,000 cells/mL, and 13.7% were BC-positive. Cathelicidin showed a strong positive correlation with SCC as demonstrated by receiver operating characteristics curve analysis and by the clustering of cathelicidin-negative and cathelicidin-positive samples in association with low and high SCC values, respectively. For evaluating the diagnostic performance of a novel test, BC cannot be considered a reliable gold standard for true disease status because of its known limitations. Therefore, we assessed the sensitivity (Se) and specificity (Sp) of the milk cathelicidin ELISA using a latent class analysis approach together with BC and SCC by considering different diagnostic thresholds to identify the preferred Se/Sp combination. We modeled conditional dependence of cathelicidin and SCC to account for their close association. The cathelicidin ELISA showed higher Se than SCC and BC for almost all threshold combinations. In fact, at the best-performing threshold combination, the Se of cathelicidin was 80.6%, 6.2 percentage points higher than that of SCC >200,000 cells/mL (74.4%) and similar to that of SCC >100,000 cells/mL (80.2%). Most importantly, this Se was obtained with a loss in Sp of only 1.4 percentage points compared with SCC >200,000 cells/mL (94.9% Sp for cathelicidin vs. 96.3% for SCC >200,000). The limited Se of BC (38.8%) was also confirmed in this study, and BC showed a slightly lower Sp than both cathelicidin and SCC for most of threshold combinations. This study confirmed that cathelicidin is released in the milk of cows with mastitis and that its presence is highly correlated with SCC. The measurement of cathelicidin by ELISA may hold significant potential for improving the sensitivity of mastitis detection in dairy cows while maintaining high specificity.
Collapse
Affiliation(s)
- M F Addis
- Porto Conte Ricerche, SP 55 Porto Conte/Capo Caccia, Km 8.400, Loc. Tramariglio, 07041 Alghero, Italy.
| | - V Tedde
- Porto Conte Ricerche, SP 55 Porto Conte/Capo Caccia, Km 8.400, Loc. Tramariglio, 07041 Alghero, Italy
| | - G M G Puggioni
- Porto Conte Ricerche, SP 55 Porto Conte/Capo Caccia, Km 8.400, Loc. Tramariglio, 07041 Alghero, Italy
| | - S Pisanu
- Porto Conte Ricerche, SP 55 Porto Conte/Capo Caccia, Km 8.400, Loc. Tramariglio, 07041 Alghero, Italy
| | - A Casula
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy
| | - C Locatelli
- Dipartimento di Scienze Veterinarie per la Salute, la Produzione Animale e la Sicurezza Alimentare, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy
| | - N Rota
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy
| | - V Bronzo
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy
| | - P Moroni
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; Animal Health Diagnostic Center, Quality Milk Production Services, Cornell University, 240 Farrier Road, Ithaca, NY 14853
| | - S Uzzau
- Porto Conte Ricerche, SP 55 Porto Conte/Capo Caccia, Km 8.400, Loc. Tramariglio, 07041 Alghero, Italy; Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Viale S. Pietro 43/B, 07100 Sassari, Italy
| |
Collapse
|
50
|
Villavicencio A, Rueda MS, Turin CG, Ochoa TJ. Factors affecting lactoferrin concentration in human milk: how much do we know? Biochem Cell Biol 2016; 95:12-21. [PMID: 28075610 DOI: 10.1139/bcb-2016-0060] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Lactoferrin (LF) is a breast milk glycoprotein with antimicrobial and anti-inflammatory effects. Its beneficial properties in infants, especially in those born preterm, are currently being studied in clinical trials. However, the maternal and nursing infant factors that may affect LF concentration in breast milk are still not clear. We conducted a systematic review to investigate the factors that may affect the concentration of LF in breast milk. We used a 2-step approach to identify the eligible studies according to inclusion/exclusion criteria, and to determine which studies would be considered. We included 70 qualified articles from 29 countries with publication dates ranging from 1976 to 2015. We described the correlation between LF concentration in breast milk and lactation stage; 10 maternal factors, such as race, parity, among others; and 2 infant factors: infections and prematurity. Colostrum has the highest LF levels, but they decrease with days postpartum. No other factor has been consistently associated with LF concentration. A major limitation of the majority of the published studies is the small sample size and the different methods used to measure LF concentration. Therefore, there is a need for large, multicenter studies with standardized study design, sample collection, and LF measurement methods to identify clinically significant factors associated with LF expression in breast milk, which will help promote exclusive breastfeeding in preterm infants.
Collapse
Affiliation(s)
- Aasith Villavicencio
- a Instituto de Medicina Tropical "Alexander von Humboldt", Universidad Peruana Cayetano Heredia, Lima 31, Peru
| | - Maria S Rueda
- a Instituto de Medicina Tropical "Alexander von Humboldt", Universidad Peruana Cayetano Heredia, Lima 31, Peru
| | - Christie G Turin
- a Instituto de Medicina Tropical "Alexander von Humboldt", Universidad Peruana Cayetano Heredia, Lima 31, Peru
| | - Theresa J Ochoa
- b Instituto de Medicina Tropical Alexander von Humboldt, Universidad Peruana Cayetano Heredia, Lima, Peru.,c University of Texas School of Public Health, Houston, TX 77030, USA
| |
Collapse
|