1
|
Puleio F, Pirri R, Tosco V, Lizio AS, Tripodi P, La Spina I, La Fauci V, Squeri R. Assessment of 2'-Fucosyllactose and Lacto-N-Neotetraose Solution as an Irrigant in E. faecalis-Infected Root Canals: An In Vitro Study. Clin Pract 2024; 14:1348-1356. [PMID: 39051302 PMCID: PMC11270313 DOI: 10.3390/clinpract14040108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/22/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Given the lack of an ideal endodontic irrigant on the market, this study evaluates the antimicrobial potential of a formulated solution of 2'-fucosyllactose and lacto-N-neotetraose against E. faecalis within infected root canals, and explores any associated impacts related to the duration of irrigation. METHODS 32 single-rooted teeth extracted for periodontal reasons were infected with Enterococcus faecalis, and subsequently subjected to endodontic treatment with two different irrigation systems: sodium hypochlorite or a solution of 2'-fucosyllactose and lacto-N-neotetraose. These samples were then incubated in sterile culture media at 37 °C to observe microbial activity through turbidity. The culture broth of each individual sample was assessed as positive or negative by observing the turbidity or lack of turbidity in the culture at the time of evaluation. RESULTS the analysis of the results obtained from the comparison of groups irrigated with sodium hypochlorite or a solution of 2'-fucosyllactose and lacto-N-neotetraose demonstrates that the case solution has no bactericidal effect against E. faecalis inoculated in the endodontic system. CONCLUSIONS the HMOs used in this study do not have a bactericidal effect on E. faecalis inoculated in an endodontic system.
Collapse
Affiliation(s)
- Francesco Puleio
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Messina University, 98100 Messina, Italy; (R.P.); (A.S.L.); (P.T.); (I.L.S.); (V.L.F.); (R.S.)
| | - Rosario Pirri
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Messina University, 98100 Messina, Italy; (R.P.); (A.S.L.); (P.T.); (I.L.S.); (V.L.F.); (R.S.)
| | - Vincenzo Tosco
- Department of Clinical Sciences and Stomatology (DISCO), Università Politecnica delle Marche, 60126 Ancona, Italy;
| | - Angelo Sergio Lizio
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Messina University, 98100 Messina, Italy; (R.P.); (A.S.L.); (P.T.); (I.L.S.); (V.L.F.); (R.S.)
| | - Paola Tripodi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Messina University, 98100 Messina, Italy; (R.P.); (A.S.L.); (P.T.); (I.L.S.); (V.L.F.); (R.S.)
| | - Isabella La Spina
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Messina University, 98100 Messina, Italy; (R.P.); (A.S.L.); (P.T.); (I.L.S.); (V.L.F.); (R.S.)
| | - Vincenza La Fauci
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Messina University, 98100 Messina, Italy; (R.P.); (A.S.L.); (P.T.); (I.L.S.); (V.L.F.); (R.S.)
| | - Raffaele Squeri
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Messina University, 98100 Messina, Italy; (R.P.); (A.S.L.); (P.T.); (I.L.S.); (V.L.F.); (R.S.)
| |
Collapse
|
2
|
Cao H, Liu Q, Liu Y, Zhao J, Qiao W, Wang Y, Liu Y, Chen L. Progress in triacylglycerol isomer detection in milk lipids. Food Chem X 2024; 22:101433. [PMID: 38764784 PMCID: PMC11101684 DOI: 10.1016/j.fochx.2024.101433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/21/2024] Open
Abstract
In triacylglycerols (TAGs), position differences of fatty acids on the glycerol skeleton produce various TAG isomers. These TAG isomers have different pathways of digestion, absorption, and utilization in infants, thereby affecting TAG nutritional properties of TAGs. Here, we review the progress of research on methods for detecting TAG isomers, and identify direction and thought for improving these methods, including novel chromatographic combinations, perfect algorithm, and improved equipment. The ensuing optimization of these methods is expected to provide robust guarantee for the gradual improvement of milk-derived TAG isomer detection, and is an important prerequisite for infant formula to mimic the structured lipids of human milk.
Collapse
Affiliation(s)
- Huiru Cao
- Key Laboratory of Dairy Science, Ministry of Education, Food Science College, Northeast Agricultural University, Harbin 150030, China
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing 100163, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing 100163, China
| | - Qian Liu
- Key Laboratory of Dairy Science, Ministry of Education, Food Science College, Northeast Agricultural University, Harbin 150030, China
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing 100163, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing 100163, China
| | - Yan Liu
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing 100163, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing 100163, China
| | - Junying Zhao
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing 100163, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing 100163, China
| | - Weicang Qiao
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing 100163, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing 100163, China
| | - Yuru Wang
- Key Laboratory of Dairy Science, Ministry of Education, Food Science College, Northeast Agricultural University, Harbin 150030, China
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing 100163, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing 100163, China
| | - Yan Liu
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing 100163, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing 100163, China
| | - Lijun Chen
- Key Laboratory of Dairy Science, Ministry of Education, Food Science College, Northeast Agricultural University, Harbin 150030, China
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing 100163, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing 100163, China
| |
Collapse
|
3
|
Marissen J, Reichert L, Härtel C, Fortmann MI, Faust K, Msanga D, Harder J, Zemlin M, Gomez de Agüero M, Masjosthusmann K, Humberg A. Antimicrobial Peptides (AMPs) and the Microbiome in Preterm Infants: Consequences and Opportunities for Future Therapeutics. Int J Mol Sci 2024; 25:6684. [PMID: 38928389 PMCID: PMC11203687 DOI: 10.3390/ijms25126684] [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: 05/10/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Antimicrobial peptides (AMPs) are crucial components of the innate immune system in various organisms, including humans. Beyond their direct antimicrobial effects, AMPs play essential roles in various physiological processes. They induce angiogenesis, promote wound healing, modulate immune responses, and serve as chemoattractants for immune cells. AMPs regulate the microbiome and combat microbial infections on the skin, lungs, and gastrointestinal tract. Produced in response to microbial signals, AMPs help maintain a balanced microbial community and provide a first line of defense against infection. In preterm infants, alterations in microbiome composition have been linked to various health outcomes, including sepsis, necrotizing enterocolitis, atopic dermatitis, and respiratory infections. Dysbiosis, or an imbalance in the microbiome, can alter AMP profiles and potentially lead to inflammation-mediated diseases such as chronic lung disease and obesity. In the following review, we summarize what is known about the vital role of AMPs as multifunctional peptides in protecting newborn infants against infections and modulating the microbiome and immune response. Understanding their roles in preterm infants and high-risk populations offers the potential for innovative approaches to disease prevention and treatment.
Collapse
Affiliation(s)
- Janina Marissen
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
- Würzburg Institute of Systems Immunology, Max-Planck Research Group, University of Würzburg, 97078 Würzburg, Germany;
| | - Lilith Reichert
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
| | - Christoph Härtel
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
- German Center for Infection Research, Site Hamburg-Lübeck-Borstel-Riems, 23538 Lübeck, Germany
| | - Mats Ingmar Fortmann
- Department of Pediatrics, University Hospital Schleswig-Holstein, 23538 Lübeck, Germany; (M.I.F.); (K.F.)
| | - Kirstin Faust
- Department of Pediatrics, University Hospital Schleswig-Holstein, 23538 Lübeck, Germany; (M.I.F.); (K.F.)
| | - Delfina Msanga
- Department of Pediatrics, Bugando Hospital, Catholic University of Health and Allied Sciences, Mwanza 33109, Tanzania;
| | - Jürgen Harder
- Department of Dermatology, Venerology and Allergology, Quincke Research Center, Kiel University, 24105 Kiel, Germany;
| | - Michael Zemlin
- Department of General Pediatrics and Neonatology, Saarland University Medical Center, 66421 Homburg, Germany;
| | - Mercedes Gomez de Agüero
- Würzburg Institute of Systems Immunology, Max-Planck Research Group, University of Würzburg, 97078 Würzburg, Germany;
| | - Katja Masjosthusmann
- Department of General Pediatrics, University Children’s Hospital Münster, 48149 Münster, Germany; (K.M.); (A.H.)
| | - Alexander Humberg
- Department of General Pediatrics, University Children’s Hospital Münster, 48149 Münster, Germany; (K.M.); (A.H.)
| |
Collapse
|
4
|
De Bernardo G, D’Urso G, Spadarella S, Giordano M, Leone G, Casapullo A. Analysis of the Fecal Metabolomic Profile in Breast vs. Different Formula Milk Feeding in Late Preterm Infants. Metabolites 2024; 14:72. [PMID: 38276307 PMCID: PMC10820811 DOI: 10.3390/metabo14010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/12/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
Human milk is the gold standard for infant nutrition, but when it is not available or insufficient to satisfy the needs of the infant, formula milk is proposed as an effective substitute. A prospective observational cohort study was conducted on late preterm infants fed with breast and two different formula milks. On this basis, they were divided into three groups: group FMPB (fed with formula + postbiotic), group FM (fed with standard formula), and group BM (breastfed). Stool samples for a metabolomic study were collected at T0 (5-7 days after birth), T1 (30 days of life), and T2 (90 days of life), giving rise to 74 samples analyzed via liquid chromatography coupled with high-resolution mass spectrometry. The T0, T1, and T2 LC-MS raw data were processed for Partial Least Square Discriminant Analysis (PLS-DA), followed by a statistical analysis. This preliminary study highlighted a good overlapping between the fecal metabolome of breast and substitute feeding systems, confirming the efficacy of the formula preparations as breast milk substitutes. Moreover, several similarities were also detected between the FMPB and BM metabolome, highlighting that the addition of a postbiotic to standard formula milk could be more effective and considered a better alternative to breast milk.
Collapse
Affiliation(s)
- Giuseppe De Bernardo
- Division of Pediatrics Neonatology and NICU, Ospedale Buon Consiglio Fatebenefratelli, 80123 Naples, Italy; (S.S.); (G.L.)
| | - Gilda D’Urso
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy;
| | - Simona Spadarella
- Division of Pediatrics Neonatology and NICU, Ospedale Buon Consiglio Fatebenefratelli, 80123 Naples, Italy; (S.S.); (G.L.)
| | - Maurizio Giordano
- Department of Clinical Medicine and Surgery, Federico II University, 80138 Naples, Italy;
| | - Giuseppina Leone
- Division of Pediatrics Neonatology and NICU, Ospedale Buon Consiglio Fatebenefratelli, 80123 Naples, Italy; (S.S.); (G.L.)
| | - Agostino Casapullo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy;
| |
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
|
Zhao S, Lok KYW, Sin ZY, Peng Y, Fan HSL, Nagesh N, Choi MSL, Kwok JYY, Choi EPH, Zhang X, Wai HKF, Tsang LCH, Cheng SSM, Wong MKL, Zhu J, Mok CKP, Ng SC, Chan FKL, Peiris M, Poon LLM, Tun HM. COVID-19 mRNA vaccine-mediated antibodies in human breast milk and their association with breast milk microbiota composition. NPJ Vaccines 2023; 8:151. [PMID: 37798293 PMCID: PMC10556030 DOI: 10.1038/s41541-023-00745-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 09/19/2023] [Indexed: 10/07/2023] Open
Abstract
Newborns can acquire immunological protection to SARS-CoV-2 through vaccine-conferred antibodies in human breast milk. However, there are some concerns around lactating mothers with regards to potential short- and long-term adverse events and vaccine-induced changes to their breast milk microbiome composition, which helps shape the early-life microbiome. Thus, we sought to explore if SARS-CoV-2 mRNA vaccine could change breast milk microbiota and how the changes impact the levels of antibodies in breast milk. We recruited 49 lactating mothers from Hong Kong who received two doses of BNT162b2 vaccine between June 2021 and August 2021. Breast milk samples were self-collected by participants pre-vaccination, one week post-first dose, one week post-second dose, and one month post-second dose. The levels of SARS-CoV-2 spike-specific IgA and IgG in breast milk peaked at one week post-second dose. Subsequently, the levels of both antibodies rapidly waned in breast milk, with IgA levels returning to baseline levels one month post-second dose. The richness and composition of human breast milk microbiota changed dynamically throughout the vaccination regimen, but the abundances of beneficial microbes such as Bifidobacterium species did not significantly change after vaccination. Additionally, we found that baseline breast milk bacterial composition can predict spike-specific IgA levels at one week post-second dose (Area Under Curve: 0.72, 95% confidence interval: 0.58-0.85). Taken together, our results identified specific breast milk microbiota markers associated with high levels of IgA in the breast milk following BNT162b2 vaccine. Furthermore, in lactating mothers, BNT162b2 vaccines did not significantly reduce probiotic species in breast milk.
Collapse
Affiliation(s)
- Shilin Zhao
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Kris Y W Lok
- School of Nursing, The University of Hong Kong, Hong Kong, China.
| | - Zhen Y Sin
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ye Peng
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Heidi S L Fan
- School of Nursing, The University of Hong Kong, Hong Kong, China
| | - Nitya Nagesh
- School of Nursing, The University of Hong Kong, Hong Kong, China
| | - Martha S L Choi
- School of Nursing, The University of Hong Kong, Hong Kong, China
| | - Jojo Y Y Kwok
- School of Nursing, The University of Hong Kong, Hong Kong, China
| | - Edmond P H Choi
- School of Nursing, The University of Hong Kong, Hong Kong, China
| | - Xi Zhang
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hogan Kok-Fung Wai
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- HKU-Pasteur Research Pole, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Leo C H Tsang
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Samuel S M Cheng
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | | | - Jie Zhu
- Microbiota I-Center (MagIC), Hong Kong SAR, China
| | - Chris K P Mok
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Siew C Ng
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Francis K L Chan
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Malik Peiris
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- HKU-Pasteur Research Pole, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Leo L M Poon
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- HKU-Pasteur Research Pole, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hein M Tun
- Microbiota I-Center (MagIC), Hong Kong SAR, China.
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
7
|
Bhattacharyya C, Barman D, Tripathi D, Dutta S, Bhattacharya C, Alam M, Choudhury P, Devi U, Mahanta J, Rasaily R, Basu A, Paine SK. Influence of Maternal Breast Milk and Vaginal Microbiome on Neonatal Gut Microbiome: a Longitudinal Study during the First Year. Microbiol Spectr 2023; 11:e0496722. [PMID: 37067419 PMCID: PMC10269640 DOI: 10.1128/spectrum.04967-22] [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: 12/02/2022] [Accepted: 03/25/2023] [Indexed: 04/18/2023] Open
Abstract
It is believed that establishment of the gut microbiome starts very early in life and is crucial for growth, immunity, and long-term metabolic health. In this longitudinal study, we recruited 25 mothers in their third trimester, of whom 15 had vaginal delivery while 10 had an unplanned cesarean section (C-section). The mother-neonate pairs were followed for 1 year, and we generated 16S metagenomic data to study the neonatal gut microbiome along with mother's breast milk and vaginal microbiomes through 12 months after delivery, at 1, 3, 6, and 12 months. We inferred (i) mode of delivery is an important factor influencing both composition and entropy of the neonatal gut microbiome, and the genus Streptococcus plays an important role in the temporal differentiation. (ii) Microbial diversity monotonically increases with age, irrespective of the mode of delivery, and it is significantly altered once exclusive breastfeeding is stopped. (iii) We found little evidence in favor of the microflora of mother's breast milk and a vaginal swab being directly reflected in the offspring's gut microbiome; however, some distinction could be made in the gut microbiome of neonates whose mothers were classified as community state type III (CSTIII) and CSTIV, based on their vaginal microbiomes. (iv) A lot of the mature gut microbiome is possibly acquired from the environment, as the genera Prevotella and Faecalibacterium, two of the most abundant flora in the neonatal gut microbiome, are introduced after initiation of solidified food. The distinction between the gut microbiome of babies born by vaginal delivery and babies born by C-section becomes blurred after introduction of solid food, although the diversity in the gut microbiota drastically increases in both cases. IMPORTANCE Gut microbiome architecture seems to have a potential impact on host metabolism, health, and nutrition. Early life gut microbiome development is considered a crucial phenomenon for neonatal health as well as adulthood metabolic complications. In this longitudinal study, we examined the association of neonatal gut microbiome entropy and its temporal variation. The study revealed that adult-like gut microbiome architecture starts taking shape after initiation of solidified food. Further, we also observed that the difference of microbial diversity was reduced between vaginally delivered and C-section babies compared to exclusive breastfeeding tenure. We found evidence in favor of the inheritance of the microflora of mother's posterior vaginal wall to the offspring's gut microbiome.
Collapse
Affiliation(s)
| | - Diganta Barman
- Department of Paediatrics, Gauhati Medical College, Assam, India
| | | | - Soumita Dutta
- Regional Medical Research Centre, Indian Council of Medical Research, Assam, India
| | - Chandra Bhattacharya
- Regional Medical Research Centre, Indian Council of Medical Research, Assam, India
| | - Mahabub Alam
- National Institute of Biomedical Genomics, West Bengal, India
| | | | - Utpala Devi
- Regional Medical Research Centre, Indian Council of Medical Research, Assam, India
| | - Jagadish Mahanta
- Regional Medical Research Centre, Indian Council of Medical Research, Assam, India
| | - Reeta Rasaily
- Maternal and Child Health, Indian Council of Medical Research, New Delhi, India
| | - Analabha Basu
- National Institute of Biomedical Genomics, West Bengal, India
| | - Suman K. Paine
- National Institute of Biomedical Genomics, West Bengal, India
- Regional Medical Research Centre, Indian Council of Medical Research, Assam, India
| |
Collapse
|
8
|
Melekoglu E, Yılmaz B, Çevik A, Gökyıldız Sürücü Ş, Avcıbay Vurgeç B, Gözüyeşil E, Sharma H, Boyan N, Ozogul F. The Impact of the Human Milk Microbiota in the Prevention of Disease and Infant Health. Breastfeed Med 2023. [PMID: 37140562 DOI: 10.1089/bfm.2022.0292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Background: Human milk is recognized as an ideal food for newborns and infants owing to the presence of various nutritive factors, including healthy bacteria. Aim/Objective: This review aimed to understand the effects of human milk microbiota in both the prevention of disease and the health of infants. Methods: Data were obtained from PubMed, Scopus, Web of Science, clinical trial registries, Dergipark, and Türk Atıf Dizini up to February 2023 without language restrictions. Results: It is considered that the first human milk microbiota ingested by the newborn creates the initial microbiome of the gut system, which in turn influences the development and maturation of immunity. Bacteria present in human milk modulate the anti-inflammatory response by releasing certain cytokines, protecting the newborn against certain infections. Therefore, certain bacterial strains isolated from human milk could serve as potential probiotics for various therapeutic applications. Conclusions: In this review, the origin and significance of human milk bacteria have been highlighted along with certain factors influencing the composition of human milk microbiota. In addition, it also summarizes the health benefits of human milk as a protective agent against certain diseases and ailments.
Collapse
Affiliation(s)
- Ebru Melekoglu
- Department of Nutrition and Dietetics, Cukurova University, Adana, Turkey
| | - Birsen Yılmaz
- Department of Nutrition and Dietetics, Cukurova University, Adana, Turkey
| | - Ayseren Çevik
- Department of Midwifery, Cukurova University, Adana, Turkey
| | | | | | - Ebru Gözüyeşil
- Department of Midwifery, Cukurova University, Adana, Turkey
| | - Heena Sharma
- Food Technology Lab, Dairy Technology Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Neslihan Boyan
- Department of Anatomy, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Fatih Ozogul
- Department of Seafood Processing Technology, Faculty of Fisheries, Cukurova University, Adana, Turkey
| |
Collapse
|
9
|
Singh DK, Miller CM, Orgel KA, Dave M, Mackay S, Good M. Necrotizing enterocolitis: Bench to bedside approaches and advancing our understanding of disease pathogenesis. Front Pediatr 2023; 10:1107404. [PMID: 36714655 PMCID: PMC9874231 DOI: 10.3389/fped.2022.1107404] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 12/20/2022] [Indexed: 01/13/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating, multifactorial disease mainly affecting the intestine of premature infants. Recent discoveries have significantly enhanced our understanding of risk factors, as well as, cellular and genetic mechanisms of this complex disease. Despite these advancements, no essential, single risk factor, nor the mechanism by which each risk factor affects NEC has been elucidated. Nonetheless, recent research indicates that maternal factors, antibiotic exposure, feeding, hypoxia, and altered gut microbiota pose a threat to the underdeveloped immunity of preterm infants. Here we review predisposing factors, status of unwarranted immune responses, and microbial pathogenesis in NEC based on currently available scientific evidence. We additionally discuss novel techniques and models used to study NEC and how this research translates from the bench to the bedside into potential treatment strategies.
Collapse
Affiliation(s)
- Dhirendra K. Singh
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Claire M. Miller
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kelly A. Orgel
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Mili Dave
- University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
| | - Stephen Mackay
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Misty Good
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
10
|
Desai H, Jones CE, Fogel JL, Negrin KA, Slater NL, Morris K, Doody LR, Engstler K, Torzone A, Smith J, Butler SC. Assessment and management of feeding difficulties for infants with complex CHD. Cardiol Young 2023; 33:1-10. [PMID: 36562257 DOI: 10.1017/s1047951122004024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Early surgical intervention in infants with complex CHD results in significant disruptions to their respiratory, gastrointestinal, and nervous systems, which are all instrumental to the development of safe and efficient oral feeding skills. Standardised assessments or treatment protocols are not currently available for this unique population, requiring the clinician to rely on knowledge based on neonatal literature. Clinicians need to be skilled at evaluating and analysing these systems to develop an appropriate treatment plan to improve oral feeding skill and safety, while considering post-operative recovery in the infant with complex CHD. Supporting the family to re-establish their parental role during the hospitalisation and upon discharge is critical to reducing parental stress and oral feeding success.
Collapse
Affiliation(s)
- Hema Desai
- Department of Rehabilitation Services, Children's Hospital of Orange County, Orange, CA, USA
| | - Courtney E Jones
- Acute Care Therapy Services, Primary Children's Hospital, Salt Lake City, UT, USA
| | - Jennifer L Fogel
- Department of Pediatric Rehabilitation, Advocate Children's Hospital, Oak Lawn, IL, USA
| | - Karli A Negrin
- Department of Therapy and Rehabilitative Services, Nemours Children's Health, Wilmington, DE, USA
| | - Nancy L Slater
- Physical Medicine and Rehabilitation Services, Children's Minnesota, Minneapolis, MN, USA
| | - Kimberly Morris
- Department of Speech-Language Pathology, Rady Children's Hospital San Diego, San Diego, CA, USA
| | - Lisa R Doody
- Pediatric Rehabilitation and Development, Advocate Children's Hospital, Oak Lawn, IL, USA
| | - Katherine Engstler
- Department of Otolaryngology and Communication Enhancement, Boston Children's Hospital, Boston, MA, USA
| | - Andrea Torzone
- Heart Center, Cardiac Intensive Care Unit, Children's Medical Center Dallas, Dallas, TX, USA
| | | | | |
Collapse
|
11
|
El-Asheer OM, El-Gazzar AF, Zakaria CM, Hassanein FA, Mohamed KA. Frequency of gastrointestinal manifestations among infants with cow’s milk protein allergy, Egypt. EGYPTIAN PEDIATRIC ASSOCIATION GAZETTE 2022. [DOI: 10.1186/s43054-022-00128-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Abstract
Background
Cow’s milk is known to be the most frequent food allergen in infants in the first years of life. Eliminating cow milk protein from diets and reintroducing it with a challenge test are the main methods for sure diagnosis. Cow’s Milk-related Symptom Score (CoMiSS) has been developed for primary health care providers to improve their knowledge about CMPA.
Objective
This study was conducted to prescribe clinical presentation of cow’s milk allergy among infants.
Methods
A cross-sectional study was conducted at Gastroenterology and Nutrition Unit, University Children Hospital, from the 1st of January 2020 to the 31st of December 2020. Forty infants, complaining of recurrent or persistent gastrointestinal manifestations, were included in our study (28 males and 12 females). Those infants were subjected to detailed medical and social patient history, comprehensive physical exam, and CoMiSS scoring as an awareness tool to suspect underlying cow milk allergy. Infants with a CoMiSS of more than 12 points and positive elimination rechallenge test were regarded as eligible to be part of our study.
Results
This study showed that the most frequently occurring gastrointestinal manifestations associated with CMPA were regurgitation (92%), diarrhea (80%), colic (75%), vomiting (67.5%), and lastly constipation which represents only (5%).
Conclusion
Regurgitation and diarrhea are the commonest presentations for infants proved to have cow milk allergy with CoMiSS score and elimination rechallenge test, where constipation is the least common presentation for those infants.
Collapse
|
12
|
Tandon P, Lee E, Jogendran R, Kroeker KI, Dieleman LA, Halloran B, Wong K, Berga KA, Huang V. Breastfeeding Patterns in Mothers with Inflammatory Bowel Disease: A Pilot Prospective Longitudinal Study. Inflamm Bowel Dis 2022; 28:1717-1724. [PMID: 35099541 DOI: 10.1093/ibd/izab354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Indexed: 12/09/2022]
Abstract
BACKGROUND Breastfeeding practices in patients with inflammatory bowel disease (IBD) remain unknown. We aimed to characterize these practices and describe factors that may lead to early discontinuation. METHODS This was a pilot, prospective, longitudinal study enrolling mothers with IBD from 2014 to 2017. Patients completed surveys on breastfeeding at time of delivery and up to 12 months postpartum. Breastfeeding discontinuation rates were reported for all patients with IBD and compared between patients with ulcerative colitis and Crohn's disease. Reproductive knowledge was defined using the Crohn's and Colitis Pregnancy Knowledge score. The Mann-Whitney U test assessed for differences between continuous variables, whereas categorical variables were compared using the chi-square test. RESULTS A total of 74 mothers with IBD were included, 47 with ulcerative colitis and 27 with Crohn's disease. Breastfeeding rates in mothers with IBD was 94.6% at delivery, 73.9% at 3 months postpartum, 55.2% at 6 months postpartum, and 30.1% at 12 months postpartum. The most common reasons for discontinuing breastfeeding before 6 months postpartum included perceived insufficient milk production and concerns of infant medication exposure through breast milk. Compared with those who continued breastfeeding beyond 6 months postpartum, those who discontinued had lower median Crohn's and Colitis Pregnancy Knowledge scores (14.0 vs 9.0; P = .04). CONCLUSIONS Though most mothers with IBD initiate breastfeeding at time of delivery, about half continue beyond 6 months postpartum. Common reasons for this include perceived insufficient milk production and medication concerns. Larger studies are required to validate our findings in more generalizable settings such as primary and secondary care.
Collapse
Affiliation(s)
- Parul Tandon
- Department of Medicine, University of Toronto, Toronto, ON, Canada.,Division of Gastroenterology and Hepatology, University of Toronto, Toronto, ON, Canada
| | - Eugenia Lee
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Rohit Jogendran
- Department of Medicine, University of Toronto, Toronto, ON, Canada.,Division of Gastroenterology and Hepatology, University of Toronto, Toronto, ON, Canada
| | - Karen I Kroeker
- Division of Gastroenterology, University of Alberta, Edmonton, AB, Canada
| | - Levinus A Dieleman
- Division of Gastroenterology, University of Alberta, Edmonton, AB, Canada
| | - Brendan Halloran
- Division of Gastroenterology, University of Alberta, Edmonton, AB, Canada
| | - Karen Wong
- Division of Gastroenterology, University of Alberta, Edmonton, AB, Canada
| | - Keri-Ann Berga
- Department of Nursing, MacEwan University, Edmonton, AB, Canada
| | - Vivian Huang
- Department of Medicine, University of Toronto, Toronto, ON, Canada.,Division of Gastroenterology and Hepatology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
13
|
Ye J, Wu Z, Zhao Y, Zhang S, Liu W, Su Y. Role of gut microbiota in the pathogenesis and treatment of diabetes mullites: Advanced research-based review. Front Microbiol 2022; 13:1029890. [PMID: 36338058 PMCID: PMC9627042 DOI: 10.3389/fmicb.2022.1029890] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 09/26/2022] [Indexed: 02/05/2023] Open
Abstract
Gut microbiota plays an important role in the proper functioning of human organisms, while its dysbiosis is associated with disease in various body organs. Diabetes mellitus (DM) is a set of heterogeneous metabolic diseases characterized by hyperglycemia caused by direct or indirect insulin deficiency. There is growing evidence that gut microbiota dysbiosis is closely linked to the development of DM. Gut microbiota composition changes in type 1 diabetes mullites (T1DM) and type 2 diabetes mullites (T2DM) patients, which may cause gut leakiness and uncontrolled entry of antigens into the circulation system, triggering an immune response that damages the isle β cells or metabolic disorders. This review summarizes gut microbiota composition in healthy individuals and compares it to diabetes mullites patients. The possible pathogenesis by which gut microbiota dysbiosis causes DM, particularly gut leakiness and changes in gut microbiota metabolites is also discussed. It also presents the process of microbial-based therapies of DM.
Collapse
Affiliation(s)
- Junjun Ye
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Shantou University Medical College, Shantou, China
| | - Zezhen Wu
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yifei Zhao
- School of Nursing, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Shuo Zhang
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Weiting Liu
- School of Nursing, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yu Su
- Center of Teaching Evaluation and Faculty Development, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
14
|
Characterization of the Metabolome of Breast Tissues from Non-Hispanic Black and Non-Hispanic White Women Reveals Correlations between Microbial Dysbiosis and Enhanced Lipid Metabolism Pathways in Triple-Negative Breast Tumors. Cancers (Basel) 2022; 14:cancers14174075. [PMID: 36077608 PMCID: PMC9454857 DOI: 10.3390/cancers14174075] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 12/02/2022] Open
Abstract
Simple Summary We previously showed that breast tumor tissues from women display an imbalance in abundance and composition of microbiota compared to normal healthy breast tissues. It is unknown whether these differences in breast tumor microbiota may be driven by alterations in microbial metabolites, leading to potentially protective or pathogenic consequences. The aim of our study was to conduct global metabolic profiling on normal and breast tumor tissues to identify differences in metabolite profiles and to determine whether breast microbial dysbiosis may be associated with enrichment of microbial metabolites in triple-negative breast cancer (TNBC) which disproportionately affects women of African ancestry. We observed significant correlations between elevated lipid metabolism pathways and microbial dysbiosis in TNBC tissues from both non-Hispanic black and white women. This is the first study to report an association between breast microbial dysbiosis and alterations in host metabolic pathways in breast tumors, including TNBC, of non-Hispanic black and non-Hispanic white women. Abstract Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer that is non-responsive to hormonal therapies and disproportionately impact women of African ancestry. We previously showed that TN breast tumors have a distinct microbial signature that differs from less aggressive breast tumor subtypes and normal breast tissues. However, it is unknown whether these differences in breast tumor microbiota may be driven by alterations in microbial metabolites, leading to potentially protective or pathogenic consequences. The goal of this global metabolomic profiling study was to investigate alterations in microbial metabolism pathways in normal and breast tumor tissues, including TNBC, of non-Hispanic black (NHB) and non-Hispanic white (NHW) women. In this study, we profiled the microbiome (16S rRNA) from breast tumor tissues and analyzed 984 metabolites from a total of 51 NHB and NHW women. Breast tumor tissues were collected from 15 patients with TNBC, 12 patients with less aggressive luminal A-type (Luminal) breast cancer, and 24 healthy controls for comparison using UHPLC-tandem mass spectrometry. Principal component analysis and hierarchical clustering of the global metabolomic profiling data revealed separation between metabolic signatures of normal and breast tumor tissues. Random forest analysis revealed a unique biochemical signature associated with elevated lipid metabolites and lower levels of microbial-derived metabolites important in controlling inflammation and immune responses in breast tumor tissues. Significant relationships between the breast microbiome and the metabolome, particularly lipid metabolism, were observed in TNBC tissues. Further investigations to determine whether alterations in sphingolipid, phospholipid, ceramide, amino acid, and energy metabolism pathways modulate Fusobacterium and Tenericutes abundance and composition to alter host metabolism in TNBC are necessary to help us understand the risk and underlying mechanisms and to identify potential microbial-based targets.
Collapse
|
15
|
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: 7.0] [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
|
16
|
Habib A, Nausheen S, Nooruddin S, Javed T, Samejo T, Hussain A, Namdev S, Amirali S, Umer M, Sheikh L, Hussain I, Ariff S, Soofi S. Effect of bovine lactoferrin on seroconversion following polio vaccine administration in children: protocol for a double-blinded randomised controlled trial. BMJ Open 2022; 12:e050849. [PMID: 35613782 PMCID: PMC9125737 DOI: 10.1136/bmjopen-2021-050849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
INTRODUCTION The oral polio vaccine (OPV) has substantial results in eliminating wild poliovirus and the vaccine of choice in polio eradication. However, the mucosal immunity induced by the OPV is still uncertain. Literature has shown that bovine lactoferrin (BLF) is a safe and useful protein found in cow's milk with extraordinary antimicrobial, antiviral, antiinflammatory and immune-modulatory functions that help children's gut to fight against micro-organisms like poliovirus. However, limited data exist regarding the effect of BLF on polio vaccine immune response. The primary objective is to evaluate the effect of BLF in enhancing mucosal and humoral immunity in children following the administration of oral and inactivated polio vaccines. METHODS AND ANALYSIS This is a two-arm double-blinded randomised controlled trial comparing 462 neonates (231 in both groups) receiving either BLF or placebo with breast milk. The intervention is administered from day 1 till 6 weeks of age to a full-term healthy singleton newborn born at the Aga Khan University Hospitals, Karachi, Pakistan. The primary outcome is the seroconversion, 1 month after the receipt of two doses of OPV (at 10 weeks). For descriptive statistical analysis, Stata will be used, the frequency with percentages will be reported to describe baseline characteristics of the participants. A χ2 test will be used to compare categorical variables and a simple t test to compare continuous variables. The proportion of seroconversion and shedding will be compared using χ2 test or Fisher's exact test. ETHICS AND DISSEMINATION The Ethics approval has been granted by the Ethics Review Committee (ERC) of Aga Khan University for the proposed trial (ID: 2019-1955-5013). Furthermore, the National Bioethics Committee (NBC) of Pakistan has also approved the study for human subject research (ID: 4-87/NBC-443/19/669). Study findings will be disseminated through presentations at scientific conferences and educational practice workshops and will be published in an international peer-reviewed scientific journal. TRIAL REGISTRATION NUMBER NCT04432935; ClinicalTrials.gov.
Collapse
Affiliation(s)
- Atif Habib
- Center of Excellence in Women & Child Health, The Aga Khan University, Karachi, Sindh, Pakistan
| | - Sidrah Nausheen
- Department of Obstetrics & Gynecology, Aga Khan University, Karachi, Sindh, Pakistan
| | - Shanila Nooruddin
- Center of Excellence in Women & Child Health, The Aga Khan University, Karachi, Sindh, Pakistan
| | - Tooba Javed
- Center of Excellence in Women & Child Health, The Aga Khan University, Karachi, Sindh, Pakistan
| | - Tariq Samejo
- Center of Excellence in Women & Child Health, The Aga Khan University, Karachi, Sindh, Pakistan
| | - Amjad Hussain
- Center of Excellence in Women & Child Health, The Aga Khan University, Karachi, Sindh, Pakistan
| | - Suneeta Namdev
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sindh, Pakistan
| | - Sehrish Amirali
- Center of Excellence in Women & Child Health, The Aga Khan University, Karachi, Sindh, Pakistan
| | - Muhammad Umer
- Center of Excellence in Women & Child Health, The Aga Khan University, Karachi, Sindh, Pakistan
| | - Lumaan Sheikh
- Department of Obstetrics & Gynecology, Aga Khan University, Karachi, Sindh, Pakistan
| | - Imtiaz Hussain
- Center of Excellence in Women & Child Health, The Aga Khan University, Karachi, Sindh, Pakistan
| | - Shabina Ariff
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sindh, Pakistan
| | - Sajid Soofi
- Center of Excellence in Women & Child Health, The Aga Khan University, Karachi, Sindh, Pakistan
- Paediatric & Child Health, The Aga Khan University, Karachi, Sindh, Pakistan
| |
Collapse
|
17
|
Millen S, Thoma-Kress AK. Milk Transmission of HTLV-1 and the Need for Innovative Prevention Strategies. Front Med (Lausanne) 2022; 9:867147. [PMID: 35360738 PMCID: PMC8962517 DOI: 10.3389/fmed.2022.867147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/18/2022] [Indexed: 11/13/2022] Open
Abstract
Breastfeeding is recommended by the World Health Organization for at least 6 months up to 2 years of age, and breast milk protects against several diseases and infections. Intriguingly, few viruses are transmitted via breastfeeding including Human T-cell leukemia virus Type 1 (HTLV-1). HTLV-1 is a highly oncogenic yet neglected retrovirus, which primarily infects CD4+ T-cells in vivo and causes incurable diseases like HTLV-1-associated inflammatory conditions or Adult T-cell leukemia/lymphoma (ATLL) after lifelong viral persistence. Worldwide, at least 5–10 million people are HTLV-1-infected and most of them are unaware of their infection posing the risk of silent transmissions. HTLV-1 is transmitted via cell-containing body fluids such as blood products, semen, and breast milk, which constitutes the major route of mother-to-child transmission (MTCT). Risk of transmission increases with the duration of breastfeeding, however, abstinence from breastfeeding as it is recommended in some endemic countries is not an option in resource-limited settings or underrepresented areas and populations. Despite significant progress in understanding details of HTLV-1 cell-to-cell transmission, it is still not fully understood, which cells in which organs get infected via the oral route, how these cells get infected, how breast milk affects this route of infection and how to inhibit oral transmission despite breastfeeding, which is an urgent need especially in underrepresented areas of the world. Here, we review these questions and provide an outlook how future research could help to uncover prevention strategies that might ultimately allow infants to benefit from breastfeeding while reducing the risk of HTLV-1 transmission.
Collapse
|
18
|
Knowledge gaps in late-onset neonatal sepsis in preterm neonates: a roadmap for future research. Pediatr Res 2022; 91:368-379. [PMID: 34497356 DOI: 10.1038/s41390-021-01721-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 12/16/2022]
Abstract
Late-onset neonatal sepsis (LONS) remains an important threat to the health of preterm neonates in the neonatal intensive care unit. Strategies to optimize care for preterm neonates with LONS are likely to improve survival and long-term neurocognitive outcomes. However, many important questions on how to improve the prevention, early detection, and therapy for LONS in preterm neonates remain unanswered. This review identifies important knowledge gaps in the management of LONS and describe possible methods and technologies that can be used to resolve these knowledge gaps. The availability of computational medicine and hypothesis-free-omics approaches give way to building bedside feedback tools to guide clinicians in personalized management of LONS. Despite advances in technology, implementation in clinical practice is largely lacking although such tools would help clinicians to optimize many aspects of the management of LONS. We outline which steps are needed to get possible research findings implemented on the neonatal intensive care unit and provide a roadmap for future research initiatives. IMPACT: This review identifies knowledge gaps in prevention, early detection, antibiotic, and additional therapy of late-onset neonatal sepsis in preterm neonates and provides a roadmap for future research efforts. Research opportunities are addressed, which could provide the means to fill knowledge gaps and the steps that need to be made before possible clinical use. Methods to personalize medicine and technologies feasible for bedside clinical use are described.
Collapse
|
19
|
Liang N, Beverly RL, Scottoline BP, Dallas DC. Peptides Derived from In Vitro and In Vivo Digestion of Human Milk Are Immunomodulatory in THP-1 Human Macrophages. J Nutr 2021; 152:331-342. [PMID: 34601601 PMCID: PMC8754566 DOI: 10.1093/jn/nxab350] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/11/2021] [Accepted: 09/23/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Milk proteins contain many encrypted bioactive peptides. Whether these bioactive peptides are released in the infant intestine and exert immunomodulatory activity remains unknown. OBJECTIVE This study examined in vitro immunomodulatory activities of peptides from in vitro- and in vivo-digested human milk. METHODS Peptides were extracted from in vitro-digested human milk and pooled intestinal samples from 8 infants fed human milk. Peptides extracted from in vitro-digested samples were fractionated. The in vitro effects of these peptides and fractions on the secretion of TNF-α and IL-8 in LPS-treated human immune THP-1 macrophages were evaluated. The significance of differences between in vitro peptide fraction treatment and control on cytokine production was analyzed by t test. LC-MS/MS-based peptidomics was conducted to identify the peptides. The peptides were screened for potential bioactivity using a sequence homology search using the Milk Bioactive Peptide Database (MBPDB). RESULTS Six fractions of the peptide mixture extracted from the in vitro-digested human milk significantly inhibited TNF-α production by LPS-challenged THP-1 macrophages. Fractions F4, F8, F11, F14, and F17 attenuated IL-8 secretion, and F6/7 and F18 increased IL-8 secretion. Peptides extracted from the pooled in vivo intestinal samples attenuated both TNF-α and IL-8 secretion. There were 266 and 418 peptides identified in the in vitro and in vivo samples, respectively. Among the peptides, 34 and 50 in the in vitro and in vivo samples, respectively, had >80% sequence similarity to bioactive peptides in the MBPDB. CONCLUSIONS Peptides released by in vitro and in vivo infant digestion of human milk were immunomodulatory in human immune cells; fractions F4, F8, and F11 were anti-inflammatory; and F6/7 and F18 were proinflammatory. Thirteen peptides were present in all fractions with anti-inflammatory activity, and 38 peptides were present in all fractions with proinflammatory activity. These peptides potentially contributed to the observed immunomodulatory activity of the peptide mixtures.
Collapse
Affiliation(s)
- Ningjian Liang
- Nutrition Program, School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, USA
| | - Robert L Beverly
- Nutrition Program, School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, USA
| | - Brian P Scottoline
- Department of Pediatrics, Oregon Health and Science University, Portland, OR, USA
| | | |
Collapse
|
20
|
Abstract
Necrotizing enterocolitis (NEC) is an inflammatory disease affecting premature infants. Intestinal microbial composition may play a key role in determining which infants are predisposed to NEC and when infants are at highest risk of developing NEC. It is unclear how to optimize antibiotic therapy in preterm infants to prevent NEC and how to optimize antibiotic regimens to treat neonates with NEC. This article discusses risk factors for NEC, how dysbiosis in preterm infants plays a role in the pathogenesis of NEC, and how probiotic and antibiotic therapy may be used to prevent and/or treat NEC and its sequelae.
Collapse
Affiliation(s)
- Jennifer Duchon
- Division of Newborn Medicine, Jack and Lucy Department of Pediatrics, Icahn School of Medicine at Mount Sinai, 1000 10th Avenue, New York, NY 10019, USA
| | - Maria E Barbian
- Division of Neonatal-Perinatal Medicine, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive Northeast, 3rd Floor, Atlanta, GA 30322, USA
| | - Patricia W Denning
- Division of Neonatal-Perinatal Medicine, Emory University School of Medicine, Children's Healthcare of Atlanta, Emory University Hospital Midtown, 550 Peachtree Street, 3rd Floor MOT, Atlanta, GA 30308, USA.
| |
Collapse
|
21
|
Cortés-Macías E, Selma-Royo M, Martínez-Costa C, Collado MC. Breastfeeding Practices Influence the Breast Milk Microbiota Depending on Pre-Gestational Maternal BMI and Weight Gain over Pregnancy. Nutrients 2021; 13:1518. [PMID: 33946343 PMCID: PMC8146841 DOI: 10.3390/nu13051518] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 01/04/2023] Open
Abstract
Breastfeeding is critical for adequate neonatal microbial and immune system development affecting neonate health outcomes in the short and long term. There is a great interest in ascertaining which are the maternal factors contributing to the milk microbiota and the potential relevance for the developing infant. Thus, our study aimed to characterize the effect of mixed and exclusive breastfeeding practices on the milk microbiota and to determine the impact of pre-pregnancy body mass index (BMI) and weight gain over pregnancy on its composition. Breast milk samples from 136 healthy women were collected within the first month post-partum and milk microbiota profiling was analyzed by 16S rRNA gene sequencing. Information on breastfeeding habits and maternal-infant clinical data were recorded. Breastfeeding practices (exclusive vs. mixed), maternal pre-gestational BMI, and weight gain over pregnancy contributed to the milk microbiota variation. Pre-gestational normal-weight women with exclusive breastfeeding habits harbored a significantly higher abundance of Bifidobacterium genus, and also, higher alpha-diversity compared to the rest of the women. Our results confirm the importance of controlling weight during pregnancy and breastfeeding practices in terms of milk microbiota. Further studies to clarify the potential impact of these maternal factors on milk and infant development and health will be necessary.
Collapse
Affiliation(s)
- Erika Cortés-Macías
- Department of Biotechnology, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), 46980 Valencia, Spain; (E.C.-M.); (M.S.-R.)
| | - Marta Selma-Royo
- Department of Biotechnology, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), 46980 Valencia, Spain; (E.C.-M.); (M.S.-R.)
| | - Cecilia Martínez-Costa
- Department of Pediatrics, INCLIVA Research Institute, School of Medicine, University of Valencia, 46003 Valencia, Spain;
- Pediatric Gastroenterology and Nutrition Section, Hospital Clínico Universitario Valencia, INCLIVA, 46010 Valencia, Spain
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), 46980 Valencia, Spain; (E.C.-M.); (M.S.-R.)
| |
Collapse
|
22
|
Grases-Pintó B, Abril-Gil M, Torres-Castro P, Castell M, Rodríguez-Lagunas MJ, Pérez-Cano FJ, Franch À. Rat Milk and Plasma Immunological Profile throughout Lactation. Nutrients 2021; 13:nu13041257. [PMID: 33920419 PMCID: PMC8070501 DOI: 10.3390/nu13041257] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/26/2021] [Accepted: 04/07/2021] [Indexed: 01/02/2023] Open
Abstract
The composition of bioactive factors with immune activity in human breast milk is widely studied. However, the knowledge on rat milk immune factors during the whole lactation period is still scarce. This study aimed to analyze rat breast milk’s immunoglobulin (Ig) content and some critical adipokines and growth factors throughout the lactation period, and to assess relationships with corresponding plasma levels. During lactation, milk concentration of the transforming growth factor (TGF)-β2 and -β3 showed a punctual increase in the first week, whereas adiponectin and leptin remained stable. In the second period of lactation (d14–21), despite the increase in the milk epidermal growth factor (EGF), a decrease in fibroblast growth factor 21 (FGF21) was detected at day 21. Milk IgA concentration had a progressive increase during lactation, while no significant changes were found in IgM and IgG. Regarding plasma levels, a decrease in all studied adipokines was observed in the second period of lactation, with the exception of IgA and TGF-β1, which reached their highest values at the end of the study. A positive correlation in IgM, IgG, and adipokine concentration was detected between milk and plasma compartments. In summary, the changes in the pattern of these bioactive compounds in rat milk and plasma and their relationships during lactation are established.
Collapse
Affiliation(s)
- Blanca Grases-Pintó
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
| | - Mar Abril-Gil
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
| | - Paulina Torres-Castro
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María J. Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
| | - Francisco J. Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
- Correspondence: ; Tel.: +34-934-024-505
| | - Àngels Franch
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (B.G.-P.); (M.A.-G.); (P.T.-C.); (M.C.); (M.J.R.-L.); (À.F.)
- Nutrition and Food Safety Research Institute (INSA·UB), 08921 Santa Coloma de Gramenet, Spain
| |
Collapse
|
23
|
Zhou Y, Yu Z, Wang X, Chen W, Liu Y, Zhang Y, Yin J, Han S. Exosomal circRNAs contribute to intestinal development via the VEGF signalling pathway in human term and preterm colostrum. Aging (Albany NY) 2021; 13:11218-11233. [PMID: 33820870 PMCID: PMC8109075 DOI: 10.18632/aging.202806] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 02/08/2021] [Indexed: 12/21/2022]
Abstract
Human breast milk (HBM) provides essential nutrients for newborn growth and development, and contains a variety of biologically active ingredients that can affect gastrointestinal tract and immune system development in breastfed infants. HBM also contains mRNAs, microRNAs and lncRNAs, most of which are encapsulated in milk-derived exosomes and exhibit various important infant development related biological functions. While previous studies have shown that exosomal circRNAs are involved in the intestinal epithelial cells’ proliferation and repair. However, the effect of HBM exosomal circRNAs on intestinal development is not clear. In this study, we identified 6756 circRNAs both in preterm colostrum (PC) and term colostrum (TC), of which 66 were upregulated, and 42 were downregulated (|fold change>2|, p < 0.05) in PC. Pathway analysis showed that the VEGF signalling pathway was involved, and network analysis revealed that the differentially expressed circRNAs bound various miRNAs. Further analyses showed that has_circRNA_405708 and has_circRNA_104707 were involved in the VEGF signalling pathway, and that they all bound various mirRNAs. Exosomes found in preterm colostrum (PC) and term colostrum (TC) promoted VEGF protein expression and induced the proliferation and migration of small intestinal epithelial cells (FHCs). Exosomal circRNAs found in human colostrum (HC) binding to related miRNAs may regulate VEGF signalling, and intestinal development.
Collapse
Affiliation(s)
- Yahui Zhou
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China.,Department of Pediatrics, Fourth Clinical Medicine College, Nanjing Medical University, Nanjing 210029, China
| | - Zhangbin Yu
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Xingyun Wang
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Wenjuan Chen
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Yiwen Liu
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Yinghui Zhang
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Jing Yin
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Shuping Han
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| |
Collapse
|
24
|
Perrella S, Gridneva Z, Lai CT, Stinson L, George A, Bilston-John S, Geddes D. Human milk composition promotes optimal infant growth, development and health. Semin Perinatol 2021; 45:151380. [PMID: 33431112 DOI: 10.1016/j.semperi.2020.151380] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human milk is a living dynamic fluid that promotes optimal nutrition and development of the infant and impacts health across the lifespan. This review reports on the diverse range of nutrients, immune protection factors, hormones, microbes and metabolites in human milk and their impacts on infant nutrition and health. While many of these components are stable across lactation and similar between women, some vary over time, and in response to maternal and infant health status, maternal diet and geographic location. Human milk may be considered as personalized nutrition, with many components working synergistically to stimulate and support the infant's immature immune system, while enhancing appropriate development, growth and body composition.
Collapse
Affiliation(s)
- Sharon Perrella
- School of Molecular Science, The University of Western Australia, M310, 35 Stirling Highway, Crawley Western Australia 6009, Australia.
| | - Zoya Gridneva
- School of Molecular Science, The University of Western Australia, M310, 35 Stirling Highway, Crawley Western Australia 6009, Australia
| | - Ching Tat Lai
- School of Molecular Science, The University of Western Australia, M310, 35 Stirling Highway, Crawley Western Australia 6009, Australia
| | - Lisa Stinson
- School of Molecular Science, The University of Western Australia, M310, 35 Stirling Highway, Crawley Western Australia 6009, Australia
| | - Alexandra George
- School of Molecular Science, The University of Western Australia, M310, 35 Stirling Highway, Crawley Western Australia 6009, Australia
| | - Sabrina Bilston-John
- School of Molecular Science, The University of Western Australia, M310, 35 Stirling Highway, Crawley Western Australia 6009, Australia
| | - Donna Geddes
- School of Molecular Science, The University of Western Australia, M310, 35 Stirling Highway, Crawley Western Australia 6009, Australia
| |
Collapse
|
25
|
Zhou H, Sun L, Zhang S, Zhao X, Gang X, Wang G. The crucial role of early-life gut microbiota in the development of type 1 diabetes. Acta Diabetol 2021; 58:249-265. [PMID: 32712802 DOI: 10.1007/s00592-020-01563-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/19/2020] [Indexed: 02/07/2023]
Abstract
Early-life healthy gut microbiota has a profound implication on shaping the mucosal immune system as well as maintaining healthy status later in life, especially at the prenatal or neonatal stages, while intestinal dysbiosis in early life is associated with several autoimmune diseases, including type 1 diabetes (T1D). Since the gut microbiome is potentially modifiable, optimizing the intestinal bacterial composition in early life may be a novel option for T1D prevention. In this review, we will review current data depicting the crucial role of early-life intestinal microbiome in the development of T1D and discuss the possible mechanisms whereby early-life intestinal microbiome influences the T1D progression. We also summarize recent findings on environmental factors affecting gut microbiota colonization and interventions that may successfully alter microbial composition to discuss potential means of preventing T1D progression in at-risk children.
Collapse
Affiliation(s)
- He Zhou
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, China
| | - Lin Sun
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, China
| | - Siwen Zhang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xue Zhao
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
26
|
Strumillo ST, Kartavykh D, de Carvalho FF, Cruz NC, de Souza Teodoro AC, Sobhie Diaz R, Curcio MF. Host-virus interaction and viral evasion. Cell Biol Int 2021; 45:1124-1147. [PMID: 33533523 PMCID: PMC8014853 DOI: 10.1002/cbin.11565] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 01/24/2021] [Indexed: 12/12/2022]
Abstract
With each infectious pandemic or outbreak, the medical community feels the need to revisit basic concepts of immunology to understand and overcome the difficult times brought about by these infections. Regarding viruses, they have historically been responsible for many deaths, and such a peculiarity occurs because they are known to be obligate intracellular parasites that depend upon the host's cell machinery for their replication. Successful infection with the production of essential viral components requires constant viral evolution as a strategy to manipulate the cellular environment, including host internal factors, the host's nonspecific and adaptive immune responses to viruses, the metabolic and energetic state of the infected cell, and changes in the intracellular redox environment during the viral infection cycle. Based on this knowledge, it is fundamental to develop new therapeutic strategies for controlling viral dissemination, by means of antiviral therapies, vaccines, or antioxidants, or by targeting the inhibition or activation of cell signaling pathways or metabolic pathways that are altered during infection. The rapid recovery of altered cellular homeostasis during viral infection is still a major challenge. Here, we review the strategies by which viruses evade the host's immune response and potential tools used to develop more specific antiviral therapies to cure, control, or prevent viral diseases.
Collapse
Affiliation(s)
- Scheilla T Strumillo
- Department of Biochemistry, Laboratory of Cell Signaling, Federal University of São Paulo, São Paulo, Brazil
| | - Denis Kartavykh
- Department of Medicine, Laboratory of Retrovirology, Federal University of São Paulo, São Paulo, Brazil
| | - Fábio F de Carvalho
- Departament of Educational Development, Getulio Vargas Foundation, São Paulo, Brazil
| | - Nicolly C Cruz
- Department of Medicine, Laboratory of Retrovirology, Federal University of São Paulo, São Paulo, Brazil
| | - Ana C de Souza Teodoro
- Department of Biochemistry, Laboratory of Cell Signaling, Federal University of São Paulo, São Paulo, Brazil
| | - Ricardo Sobhie Diaz
- Department of Medicine, Laboratory of Retrovirology, Federal University of São Paulo, São Paulo, Brazil
| | - Marli F Curcio
- Department of Medicine, Laboratory of Retrovirology, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
27
|
Kendall E, Millard A, Beaumont J. The "weanling's dilemma" revisited: Evolving bodies of evidence and the problem of infant paleodietary interpretation. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2021; 175 Suppl 72:57-78. [PMID: 33460467 DOI: 10.1002/ajpa.24207] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 10/23/2020] [Accepted: 12/06/2020] [Indexed: 01/02/2023]
Abstract
Breastfeeding is known to be a powerful mediator of maternal and childhood health, with impacts throughout the life course. Paleodietary studies of the past 30 years have accordingly taken an enduring interest in the health and diet of young children as a potential indicator of population fertility, subsistence, and mortality patterns. While progress has been made in recent decades toward acknowledging the agency of children, many paleodietary reconstructions have failed to incorporate developments in cognate disciplines revealing synergistic dynamics between maternal and offspring biology. Paleodietary interpretation has relied heavily on the "weanling's dilemma," in which infants are thought to face a bleak choice between loss of immunity or malnutrition. Using a review of immunological and epidemiological evidence for the dynamic and supportive role that breastfeeding plays throughout the complementary feeding period, this article offers context and nuance for understanding past feeding transitions. We suggest that future interpretative frameworks for infant paleodietary and bioarchaeological research should include a broad knowledge base that keeps pace with relevant developments outside of those disciplines.
Collapse
Affiliation(s)
- Ellen Kendall
- Department of Archaeology, Durham University, Durham, UK
| | - Andrew Millard
- Department of Archaeology, Durham University, Durham, UK
| | - Julia Beaumont
- School of Archaeological and Forensic Sciences, University of Bradford, Bradford, UK
| |
Collapse
|
28
|
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: 5] [Impact Index Per Article: 1.3] [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
|
29
|
Nordenström K, Lannering K, Mellander M, Elfvin A. Low risk of necrotising enterocolitis in enterally fed neonates with critical heart disease: an observational study. Arch Dis Child Fetal Neonatal Ed 2020; 105:609-614. [PMID: 32170030 PMCID: PMC7592358 DOI: 10.1136/archdischild-2019-318537] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 02/13/2020] [Accepted: 02/20/2020] [Indexed: 01/23/2023]
Abstract
OBJECTIVE We aimed to investigate the frequency of necrotising enterocolitis (NEC) in infants with critical congenital heart disease (CCHD) hypothesising that preoperative enteral feeding does not increase the risk of NEC. BACKGROUND When NEC affects term infants, underlying risk factors such as asphyxia, sepsis or CCHD are often found. Due to fear of NEC development in infants with CCHD great caution is practised in many countries to defer preoperative enteral feeding, but in Sweden this is routinely provided. DESIGN, SETTING AND PATIENTS An observational study of all infants born with CCHD who were admitted to Queen Silvia Children's Hospital in Gothenburg between 2010 and 2017. The International Classification of Diseases 10th Revision diagnosis code of NEC was used to identify NEC cases in this group. Infants described as 'fully fed' or who were fed at least 45 mL/kg/day before cardiac surgery were identified. MAIN OUTCOME MEASURES NEC in infants with CCHD in relation to preoperative enteral feeding. RESULTS There were 458 infants with CCHD admitted during the study period. 408/458 were born at term and 361/458 required prostaglandin E1 before surgery. In total, 444/458 infants (97%) were fully fed or fed at least 45 mL/kg daily before cardiac surgery. Four of 458 infants developed NEC (0.9%). All four had other risk factors for NEC. CONCLUSIONS This study showed a low risk of NEC in term infants fed enterally before cardiac surgery. We speculate that preoperative enteral feeding of neonates with CCHD does not increase the risk of NEC development.
Collapse
Affiliation(s)
- Kajsa Nordenström
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, Goteborg, Sweden,Department of Pediatrics, Sahlgrenska University Hospital, Goteborg, Sweden
| | - Katarina Lannering
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, Goteborg, Sweden,Pediatric Heart Center, Sahlgrenska University Hospital, Goteborg, Sweden
| | - Mats Mellander
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, Goteborg, Sweden,Pediatric Heart Center, Sahlgrenska University Hospital, Goteborg, Sweden
| | - Anders Elfvin
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, Goteborg, Sweden .,Department of Pediatrics, Sahlgrenska University Hospital, Goteborg, Sweden
| |
Collapse
|
30
|
Blends of Human Milk Oligosaccharides Confer Intestinal Epithelial Barrier Protection in Vitro. Nutrients 2020; 12:nu12103047. [PMID: 33027993 PMCID: PMC7599875 DOI: 10.3390/nu12103047] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/29/2020] [Accepted: 10/02/2020] [Indexed: 12/31/2022] Open
Abstract
Breastfeeding is integral in the proper maturation of the intestinal barrier and protection against inflammatory diseases. When human milk (HM) is not available, supplementation with HM bioactives like Human Milk Oligosaccharides (HMOs) may help in providing breastfeeding barrier-protective benefits. An increasing HMO variety is becoming industrially available, enabling approaching the HMO complexity in HM. We aimed at assessing the impact of blends of available HMOs on epithelial barrier function in vitro. The capacity of individual [2'-Fucosyllactose (2'FL), Difucosyllactose, Lacto-N-tetraose, Lacto-N-neotetraose, 3'-Siallylactose and 6'-Siallylactose] or varying combinations of 3, 5 and 6 HMOs to modulate fluorescein-isothiocyanate (FITC)-labelled Dextran 4 KDa (FD4) translocation and/or transepithelial resistance (TEER) was characterized in Caco-2: HT29- methotrexate (MTX) cell line monolayers before and after an inflammatory challenge with TNF-α and IFN-γ. The six HMO blend (HMO6) dose-dependently limited the cytokine-induced FD4 translocation and TEER decrease and increased TEER values before challenge. Similarly, 3 and 5 HMO blends conferred a significant protection against the challenge, with 2'FL, one of the most abundant but most variable oligosaccharides in HM, being a key contributor. Overall, our results suggest differential ability of specific HMOs in modulating the intestinal barrier and support the potential of supplementation with combinations of available HMOs to promote gut health and protect against intestinal inflammatory disorders.
Collapse
|
31
|
JANSEN SUSIANA, WASITYASTUTI WIDYA, ASTARINI FAJARDWI, HARTINI SRI. Mothers' knowledge of breastfeeding and infant feeding types affect acute respiratory infections. JOURNAL OF PREVENTIVE MEDICINE AND HYGIENE 2020; 61:E401-E408. [PMID: 33150229 PMCID: PMC7595061 DOI: 10.15167/2421-4248/jpmh2020.61.3.1499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/15/2020] [Indexed: 01/12/2023]
Abstract
Introduction World Health Organization (WHO) recommends exclusive breastfeeding for new-borns until 6 months of age. However, exclusive breastfeeding in Indonesia only reached 52.3% in 2014 and 65.16% in 2018. It is known that administration of infant formula and non-formula supplements to infants aged less than 6 months increases the risk of Acute Respiratory Infections (ARIs). In addition, the high prevalence of ARIs in infants in Sleman Regency, Indonesia indicates the need of optimal early prevention. Therefore, we conducted this study to confirm that mothers’ knowledge of breastfeeding and infant feeding types affect the prevalence of Acute Respiratory Infections (ARIs). Methods Data were collected through questionnaires from 50 mothers with infants aged 7-12 months who had experienced ARIs in the last 3 months (case group) and 50 mothers with healthy infants (control group). Collected data were then analysed using Chi-Square, Logistic Regression, Lambda, and Somers’ D tests. Results The results showed that types of infant feeding are associated with the prevalence of ARIs. Non-breastfed infants were 14 times riskier to contract ARIs. Mothers’ knowledge of exclusive breastfeeding influenced their preferences of feeding practice. However, their attitude towards breastfeeding did not appear to significantly affect their choices of feeding practice. Conclusions Exclusive breastfeeding during the first 6 months of an infant’s life can lower the prevalence of ARIs for when they are older. Mothers’ good knowledge of breastfeeding is associated with its practice.
Collapse
Affiliation(s)
- SUSIANA JANSEN
- PELNI Nursing Academy of Jakarta, Indonesia
- Master in Nursing Program, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Indonesia
| | - WIDYA WASITYASTUTI
- Department of Physiology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Indonesia
- Correspondence: Widya Wasityastuti, Department of Physiology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Jl. Farmako, Senolowo, Sekip Utara, Mlati, Kabupaten Sleman, Daerah Istimewa Yogyakarta 55281, Indonesia - Tel. +62-274-6492492 - Fax. +62-274-631185 - E-mail:
| | - FAJAR DWI ASTARINI
- Master in Biomedical Sciences, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Indonesia
| | - SRI HARTINI
- Department of Pediatric Nursing and Maternity, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Indonesia
| |
Collapse
|
32
|
Sah BNP, Lueangsakulthai J, Kim BJ, Hauser BR, Woo Y, Olyaei A, Aloia M, O'Connor A, Scottoline B, Pastey MK, Dallas DC. Partial Degradation of Recombinant Antibody Functional Activity During Infant Gastrointestinal Digestion: Implications for Oral Antibody Supplementation. Front Nutr 2020; 7:130. [PMID: 32923453 PMCID: PMC7456970 DOI: 10.3389/fnut.2020.00130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 07/06/2020] [Indexed: 12/27/2022] Open
Abstract
Oral administration of engineered immunoglobulins has the potential to prevent enteric pathogen-induced diarrhea in infants. To prevent infection, these antibodies need to survive functionally intact in the proteolytic environment of the gastrointestinal tract. This research examined both ex vivo and in vivo the functional survival across infant digestion of palivizumab, a model FDA-approved recombinant antibody against respiratory syncytial virus (RSV) F protein. Palivizumab-fortified feed (formula or human milk), infant gastric, and intestinal samples were incubated to simulate in vivo digestion (ex vivo digestion). Palivizumab-fortified human milk was also fed to infants, followed by collection of gastric and intestinal samples (in vivo digestion). Palivizumab was purified from the samples of digestate using protein G spin columns followed by filtration through molecular weight cut-off membranes (30 kDa). Palivizumab functional survival across ex vivo and in vivo digestion was determined via an anti-idiotype ELISA and an RSV plaque reduction neutralization test. Palivizumab concentration and RSV neutralization capacity both decreased when incubated in intestinal samples (ex vivo study). The concentration and neutralization activity of orally-supplemented palivizumab also decreased across infant digestion (in vivo study). These results indicate that if recombinant IgGs were selected for oral supplementation to prevent enteric infections, appropriate dosing would need to account for degradation occurring in the digestive system. Other antibody formats, structural changes, or encapsulation could enhance survival in the infant gastrointestinal tract.
Collapse
Affiliation(s)
- Baidya Nath P Sah
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, United States
| | - Jiraporn Lueangsakulthai
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, United States
| | - Bum Jin Kim
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, United States
| | - Benjamin R Hauser
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, United States
| | - Yeonhee Woo
- Division of Neonatology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Amy Olyaei
- Division of Neonatology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Molly Aloia
- Division of Neonatology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Ann O'Connor
- Division of Neonatology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Brian Scottoline
- Division of Neonatology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Manoj K Pastey
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, United States
| | - David C Dallas
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, United States
| |
Collapse
|
33
|
Huang N, Wang M, Peng J, Wei H. Role of arachidonic acid-derived eicosanoids in intestinal innate immunity. Crit Rev Food Sci Nutr 2020; 61:2399-2410. [PMID: 32662287 DOI: 10.1080/10408398.2020.1777932] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Arachidonic acid (ARA), an n-6 essential fatty acid, plays an important role in human and animal growth and development. The ARA presents in the membrane phospholipids can be released by phospholipase A2. These free arachidonic acid molecules are then used to produce eicosanoids through three different pathways. Previous studies have demonstrated that eicosanoids have a wide range of physiological functions. Although they are generally considered to be pro-inflammatory molecules, recent advances have elucidated they have an effect on innate immunity via regulating the development, and differentiation of innate immune cells and the function of the intestinal epithelial barrier. Here, we review eicosanoids generation in intestine and their role in intestinal innate immunity, focusing on intestinal epithelial barrier, innate immune cell in lamina propria (LP) and their crosstalk.
Collapse
Affiliation(s)
- Ningning Huang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Miaomiao Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| |
Collapse
|
34
|
Analysis of immune, microbiota and metabolome maturation in infants in a clinical trial of Lactobacillus paracasei CBA L74-fermented formula. Nat Commun 2020; 11:2703. [PMID: 32483147 PMCID: PMC7264213 DOI: 10.1038/s41467-020-16582-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/05/2020] [Indexed: 12/15/2022] Open
Abstract
Mother’s milk is the best choice for infants nutrition, however when it is not available or insufficient to satisfy the needs of the infant, formula is proposed as an effective substitute. Here, we report the results of a randomized controlled clinical trial (NCT03637894) designed to evaluate the effects of two different dietary regimens (standard formula and Lactobacillus paracasei CBA L74-fermented formula) versus breastfeeding (reference group) on immune defense mechanisms (primary endpoint: secretory IgA, antimicrobial peptides), the microbiota and its metabolome (secondary outcomes), in healthy full term infants according to the type of delivery (n = 13/group). We show that the fermented formula, safe and well tolerated, induces an increase in secretory IgA (but not in antimicrobial peptides) and reduces the diversity of the microbiota, similarly, but not as much as, breastmilk. Metabolome analysis allowed us to distinguish subjects based on their dietary regimen and mode of delivery. Together, these results suggest that a fermented formula favors the maturation of the immune system, microbiota and metabolome. Milk breastfeeding and prebiotic-supplemented formulas have varying effects on the infant gut microbiome. Here, in a randomized controlled clinical trial, the authors investigate the effects of a Lactobacillus paracasei-fermented formula on the immune defense mechanisms, microbiota and its metabolome in full term infants.
Collapse
|
35
|
Hu P, Zhao F, Wang J, Zhu W. Early-life lactoferrin intervention modulates the colonic microbiota, colonic microbial metabolites and intestinal function in suckling piglets. Appl Microbiol Biotechnol 2020; 104:6185-6197. [DOI: 10.1007/s00253-020-10675-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/22/2020] [Accepted: 05/10/2020] [Indexed: 12/13/2022]
|
36
|
Sampah MES, Hackam DJ. Dysregulated Mucosal Immunity and Associated Pathogeneses in Preterm Neonates. Front Immunol 2020; 11:899. [PMID: 32499778 PMCID: PMC7243348 DOI: 10.3389/fimmu.2020.00899] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 04/17/2020] [Indexed: 12/12/2022] Open
Abstract
Many functions of the immune system are impaired in neonates, allowing vulnerability to serious bacterial, viral and fungal infections which would otherwise not be pathogenic to mature individuals. This vulnerability is exacerbated in compromised newborns such as premature neonates and those who have undergone surgery or who require care in an intensive care unit. Higher susceptibility of preterm neonates to infections is associated with delayed immune system maturation, with deficiencies present in both the innate and adaptive immune components. Here, we review recent insights into early life immunity, and highlight features associated with compromised newborns, given the challenges of studying neonatal immunity in compromised neonates due to the transient nature of this period of life, and logistical and ethical obstacles posed by undertaking studies newborns and infants. Finally, we highlight how the unique immunological characteristics of the premature host play key roles in the pathogenesis of diseases that are unique to this population, including necrotizing enterocolitis and the associated sequalae of lung and brain injury.
Collapse
Affiliation(s)
- Maame Efua S Sampah
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - David J Hackam
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
37
|
Trinta VDO, Padilha PDC, Petronilho S, Santelli RE, Braz BF, Freire AS, Saunders C, Rocha HFD, Sanz-Medel A, Fernández-Sánchez ML. Total metal content and chemical speciation analysis of iron, copper, zinc and iodine in human breast milk using high-performance liquid chromatography separation and inductively coupled plasma mass spectrometry detection. Food Chem 2020; 326:126978. [PMID: 32413760 DOI: 10.1016/j.foodchem.2020.126978] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 05/01/2020] [Accepted: 05/03/2020] [Indexed: 02/08/2023]
Abstract
The aim of this research was to quantify essential trace elements (iron, copper, zinc and iodine) and establish their speciation in human milk. Both the element and the species are important in new-born nutrition. Colostrum, and transitional and mature milks (25) were collected from 18 mothers of pre-term or full-term infants. Concentrations of the target elements were determined using ICP-MS. For speciation, HPLC coupled to ICP-MS was employed. Total contents of the micronutrients varied in mothers of pre-term (Fe = 0.997, Cu = 0.506, Zn = 4.15 and I = 0.458 mg L-1) and mothers of full-term (Fe = 0.733, Cu = 0.234, Zn = 2.91 and I = 0.255 mg L-1) infants. Fe, Cu and Zn were associated with biomolecules with high molecular mass compounds, such as immunoglobulins, albumin and lactoferrin whilst iodine was only found as iodide.
Collapse
Affiliation(s)
- Vânia de Oliveira Trinta
- Maternity School of the Federal University of Rio de Janeiro, Professional Master's Program in Perinatal Health, Rua das Laranjeiras, 180, CEP 22.240-000 Rio de Janeiro, Brazil
| | - Patrícia de Carvalho Padilha
- Maternity School of the Federal University of Rio de Janeiro, Professional Master's Program in Perinatal Health, Rua das Laranjeiras, 180, CEP 22.240-000 Rio de Janeiro, Brazil; Josué de Castro Nutrition Institute, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, 373, CCS, Bloco J, 2° andar, Cidade Universitária, CEP 21941-902 Rio de Janeiro, Brazil
| | - Sonaly Petronilho
- Maternity School of the Federal University of Rio de Janeiro, Professional Master's Program in Perinatal Health, Rua das Laranjeiras, 180, CEP 22.240-000 Rio de Janeiro, Brazil
| | - Ricardo Erthal Santelli
- Analytical Chemistry Department, Federal University of Rio de Janeiro, Av. Athos da Silveira Ramos 149, CT, Bloco A, 5° andar, Cidade Universitária, CEP 21941-909 Rio de Janeiro, Brazil; National Institute of Science and Technology for Bioanalytics - INCTBio, CEP 13083-970 Campinas, Brazil.
| | - Bernardo Ferreira Braz
- Analytical Chemistry Department, Federal University of Rio de Janeiro, Av. Athos da Silveira Ramos 149, CT, Bloco A, 5° andar, Cidade Universitária, CEP 21941-909 Rio de Janeiro, Brazil; National Institute of Science and Technology for Bioanalytics - INCTBio, CEP 13083-970 Campinas, Brazil
| | - Aline Soares Freire
- Analytical Chemistry Department, Federal University of Rio de Janeiro, Av. Athos da Silveira Ramos 149, CT, Bloco A, 5° andar, Cidade Universitária, CEP 21941-909 Rio de Janeiro, Brazil; National Institute of Science and Technology for Bioanalytics - INCTBio, CEP 13083-970 Campinas, Brazil
| | - Cláudia Saunders
- Maternity School of the Federal University of Rio de Janeiro, Professional Master's Program in Perinatal Health, Rua das Laranjeiras, 180, CEP 22.240-000 Rio de Janeiro, Brazil; Josué de Castro Nutrition Institute, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, 373, CCS, Bloco J, 2° andar, Cidade Universitária, CEP 21941-902 Rio de Janeiro, Brazil
| | - Hélio Fernandes da Rocha
- Medical School, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, 373, CCS, Bloco K, 2° andar, Cidade Universitária, CEP 21941-902 Rio de Janeiro, Brazil
| | - Alfredo Sanz-Medel
- Physical and Analytical Chemistry Department, Faculty of Chemistry, University of Oviedo, Oviedo, Spain
| | | |
Collapse
|
38
|
Understanding the Elements of Maternal Protection from Systemic Bacterial Infections during Early Life. Nutrients 2020; 12:nu12041045. [PMID: 32290170 PMCID: PMC7230816 DOI: 10.3390/nu12041045] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 02/06/2023] Open
Abstract
Late-onset sepsis (LOS) and other systemic bloodstream infections are notable causes of neonatal mortality, particularly in prematurely born very low birth weight infants. Breastfeeding in early life has numerous health benefits, impacting the health of the newborn in both the short-term and in the long-term. Though the known benefits of an exclusive mother's own milk diet in early life have been well recognized and described, it is less understood how breastfed infants enjoy a potential reduction in risk of LOS and other systemic infections. Here we review how gut residing pathogens within the intestinal microbiota of infants can cause a subset of sepsis cases and the components of breastmilk that may prevent the dissemination of pathogens from the intestine.
Collapse
|
39
|
Nuclear Magnetic Resonance Metabolomics Reveals Qualitative and Quantitative Differences in the Composition of Human Breast Milk and Milk Formulas. Nutrients 2020; 12:nu12040921. [PMID: 32230787 PMCID: PMC7230615 DOI: 10.3390/nu12040921] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 01/21/2023] Open
Abstract
Commercial formula milk (FM) constitutes the best alternative to fulfill the nutritional requirements of infants when breastfeeding is precluded. Here, we present the comparative study of polar metabolite composition of human breast milk (HBM) and seven different brands of FM by nuclear magnetic resonance spectroscopy. The results of the multivariate data analysis exposed qualitative and quantitative differences between HBM and FM composition as well as within FM of various brands and in HBM itself (between individual mothers and lactation period). Several metabolites were found exclusively in HBM and FM. Statistically significant higher levels of isoleucine and methionine in their free form were detected in FM samples based on caprine milk, while FM samples based on bovine milk showed a higher level of glucose and galactose in comparison to HBM. The results suggest that the amelioration of FM formulation is imperative to better mimic the composition of minor nutrients in HBM.
Collapse
|
40
|
Lueangsakulthai J, Sah BNP, Scottoline BP, Dallas DC. Survival of Recombinant Monoclonal Antibodies (IgG, IgA and sIgA) Versus Naturally-Occurring Antibodies (IgG and sIgA/IgA) in an Ex Vivo Infant Digestion Model. Nutrients 2020; 12:E621. [PMID: 32120792 PMCID: PMC7146391 DOI: 10.3390/nu12030621] [Citation(s) in RCA: 11] [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: 01/06/2020] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 01/03/2023] Open
Abstract
To prevent infectious diarrhea in infants, orally-supplemented enteric pathogen-specific recombinant antibodies would need to resist degradation in the gastrointestinal tract. Palivizumab, a recombinant antibody specific to respiratory syncytial virus (RSV), was used as a model to assess the digestion of neutralizing antibodies in infant digestion. The aim was to determine the remaining binding activity of RSV F protein-specific monoclonal and naturally-occurring immunoglobulins (Ig) in different isoforms (IgG, IgA, and sIgA) across an ex vivo model of infant digestion. RSV F protein-specific monoclonal immunoglobulins (IgG, IgA, and sIgA) and milk-derived naturally-occurring Ig (IgG and sIgA/IgA) were exposed to an ex vivo model of digestion using digestive samples from infants (gastric and intestinal samples). The survival of each antibody was tested via an RSV F protein-specific ELISA. Ex vivo gastric and intestinal digestion degraded palivizumab IgG, IgA, and sIgA (p < 0.05). However, the naturally-occurring RSV F protein-specific IgG and sIgA/IgA found in human milk were stable across gastric and intestinal ex vivo digestion. The structural differences between recombinant and naturally-occurring antibodies need to be closely examined to guide future design of recombinant antibodies with increased stability for use in the gastrointestinal tract.
Collapse
Affiliation(s)
- Jiraporn Lueangsakulthai
- Nutrition Program, School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR 97331, USA; (J.L.); (B.N.P.S.)
| | - Baidya Nath P. Sah
- Nutrition Program, School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR 97331, USA; (J.L.); (B.N.P.S.)
| | - Brian P. Scottoline
- Department of Pediatrics, Oregon Health and Sciences University, Portland, OR 97239, USA;
| | - David C. Dallas
- Nutrition Program, School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR 97331, USA; (J.L.); (B.N.P.S.)
| |
Collapse
|
41
|
Rodríguez-Camejo C, Puyol A, Fazio L, Villamil E, Arbildi P, Sóñora C, Castro M, Carroscia L, Hernández A. Impact of Holder pasteurization on immunological properties of human breast milk over the first year of lactation. Pediatr Res 2020; 87:32-41. [PMID: 31288249 DOI: 10.1038/s41390-019-0500-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/26/2019] [Accepted: 07/02/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND The timing of milk donations to human milk banks ranges from a few days to more than 1 year after delivery, and the Holder method is used for pasteurization. We evaluated the effect of temporal variation and thermal treatment on the immunological properties of milk. METHODS We analyzed 73 milk samples, raw and after pasteurization, donated at different lactation stages. We studied antibodies, lysozyme, cytokines, soluble receptors, and factors with impact on barrier function. We also evaluated in vitro the capacity of milk to modulate nuclear factor-κB (NF-κB) signaling in an HT-29 epithelial cell line stimulated with tumor necrosis factor-α (TNF-α). RESULTS With few exceptions, immune components exhibited their highest levels in colostrum, and were stable in the various stages of mature milk. Pasteurization altered the immunological composition of milk, and very drastically for some components. Raw milk of the first year reduced NF-κB activation in HT-29 cells treated with TNF-α to approximately the same extent, and Holder pasteurization significantly affected this capacity. CONCLUSIONS Overall, the present work reports that mature donated milk is equally valuable over the first year of lactation, but warns about drastic losses of anti-inflammatory properties during Holder pasteurization that could be critical for the health of preterm infants.
Collapse
Affiliation(s)
- Claudio Rodríguez-Camejo
- Cátedra de Inmunología, Instituto de Química Biológica, Facultad de Ciencias, Área Inmunología, Departamento de Biociencias, Facultad de Química, Instituto de Higiene, Universidad de la República, Montevideo, Uruguay
| | - Arturo Puyol
- Banco de Leche "Ruben Panizza", Centro Hospitalario Pereira Rossell, Administración de los Servicios de Salud del Estado, Montevideo, Uruguay
| | - Laura Fazio
- Banco de Leche "Ruben Panizza", Centro Hospitalario Pereira Rossell, Administración de los Servicios de Salud del Estado, Montevideo, Uruguay
| | - Emilia Villamil
- Cátedra de Inmunología, Instituto de Química Biológica, Facultad de Ciencias, Área Inmunología, Departamento de Biociencias, Facultad de Química, Instituto de Higiene, Universidad de la República, Montevideo, Uruguay
| | - Paula Arbildi
- Cátedra de Inmunología, Instituto de Química Biológica, Facultad de Ciencias, Área Inmunología, Departamento de Biociencias, Facultad de Química, Instituto de Higiene, Universidad de la República, Montevideo, Uruguay
| | - Cecilia Sóñora
- Cátedra de Inmunología, Instituto de Química Biológica, Facultad de Ciencias, Área Inmunología, Departamento de Biociencias, Facultad de Química, Instituto de Higiene, Universidad de la República, Montevideo, Uruguay.,Escuela Universitaria de Tecnología Médica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Mara Castro
- Hospital de la Mujer, Centro Hospitalario Pereira Rossell, Administración de los Servicios de Salud del Estado, Montevideo, Uruguay
| | - Lilian Carroscia
- Banco de Leche "Ruben Panizza", Centro Hospitalario Pereira Rossell, Administración de los Servicios de Salud del Estado, Montevideo, Uruguay
| | - Ana Hernández
- Cátedra de Inmunología, Instituto de Química Biológica, Facultad de Ciencias, Área Inmunología, Departamento de Biociencias, Facultad de Química, Instituto de Higiene, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
42
|
Enman MA, Wilkinson LT, Meloni KB, Shroyer MC, Jackson TF, Aban I, Dimmitt RA, Martin CA, Galloway DP. Key Determinants for Achieving Enteral Autonomy and Reduced Parenteral Nutrition Exposure in Pediatric Intestinal Failure. JPEN J Parenter Enteral Nutr 2019; 44:1263-1270. [DOI: 10.1002/jpen.1754] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 11/11/2019] [Indexed: 01/08/2023]
Affiliation(s)
- Macie A. Enman
- University of Alabama at Birmingham School of Medicine Birmingham Alabama USA
| | - Linda T. Wilkinson
- Division of Pediatric Gastroenterology Hepatology, & Nutrition University of Alabama at Birmingham School of Medicine Birmingham Alabama USA
| | - Katie B. Meloni
- Deparment of Clinical Nutrition Children's of Alabama Birmingham Alabama USA
| | - Michelle C. Shroyer
- Division of Pediatric Surgery University of Alabama at Birmingham Birmingham Alabama USA
| | - Tracy F. Jackson
- Department of Pharmacy Children's of Alabama Birmingham Alabama USA
| | - Inmaculada Aban
- Department of Biostatistics School of Public Health University of Alabama at Birmingham Birmingham Alabama USA
| | - Reed A. Dimmitt
- Division of Pediatric Gastroenterology Hepatology, & Nutrition University of Alabama at Birmingham School of Medicine Birmingham Alabama USA
| | - Colin A. Martin
- Division of Pediatric Surgery University of Alabama at Birmingham Birmingham Alabama USA
| | - David P. Galloway
- Division of Pediatric Gastroenterology Hepatology, & Nutrition University of Alabama at Birmingham School of Medicine Birmingham Alabama USA
| |
Collapse
|
43
|
Transporters in the Mammary Gland-Contribution to Presence of Nutrients and Drugs into Milk. Nutrients 2019; 11:nu11102372. [PMID: 31590349 PMCID: PMC6836069 DOI: 10.3390/nu11102372] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/19/2019] [Accepted: 09/25/2019] [Indexed: 02/07/2023] Open
Abstract
A large number of nutrients and bioactive ingredients found in milk play an important role in the nourishment of breast-fed infants and dairy consumers. Some of these ingredients include physiologically relevant compounds such as vitamins, peptides, neuroactive compounds and hormones. Conversely, milk may contain substances-drugs, pesticides, carcinogens, environmental pollutants-which have undesirable effects on health. The transfer of these compounds into milk is unavoidably linked to the function of transport proteins. Expression of transporters belonging to the ATP-binding cassette (ABC-) and Solute Carrier (SLC-) superfamilies varies with the lactation stages of the mammary gland. In particular, Organic Anion Transporting Polypeptides 1A2 (OATP1A2) and 2B1 (OATP2B1), Organic Cation Transporter 1 (OCT1), Novel Organic Cation Transporter 1 (OCTN1), Concentrative Nucleoside Transporters 1, 2 and 3 (CNT1, CNT2 and CNT3), Peptide Transporter 2 (PEPT2), Sodium-dependent Vitamin C Transporter 2 (SVCT2), Multidrug Resistance-associated Protein 5 (ABCC5) and Breast Cancer Resistance Protein (ABCG2) are highly induced during lactation. This review will focus on these transporters overexpressed during lactation and their role in the transfer of products into the milk, including both beneficial and harmful compounds. Furthermore, additional factors, such as regulation, polymorphisms or drug-drug interactions will be described.
Collapse
|
44
|
Soluble CD14 in Breast Milk and Its Relation to Atopic Manifestations in Early Infancy. Nutrients 2019; 11:nu11092118. [PMID: 31492016 PMCID: PMC6770418 DOI: 10.3390/nu11092118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/29/2019] [Accepted: 08/30/2019] [Indexed: 12/15/2022] Open
Abstract
Soluble CD14 (sCD14) is one of the immunomodulatory factors in breast milk (BM). Although it may be involved in the prevention of atopic symptoms and sensitization to both food and inhalant allergens, conflicting evidence exists concerning its protective effects. In this study, we investigated the relationship between sCD14 in colostrum and 1-month BM, and the development of atopic dermatitis (AD) and sensitization to food and aeroallergens at 9 months of age in infants who were exclusively or almost exclusively breastfed up to 4 months of age. BM samples were collected from lactating mothers who participated in a 2 × 2 factorial, randomized, nontreatment controlled trial study set in Tokyo, which looked at the efficacy of emollients and synbiotics in preventing AD and food allergy in children during the first year of life. A total of 258 colostrum samples and 269 1-month BM samples were analyzed. We found that one-month BM sCD14 levels in the AD group were significantly lower than in the non-AD group. Levels of sCD14 in 1-month BM were not related to allergen sensitization in the overall analysis, but egg white sensitization correlated inversely with 1-month BM sCD14 in infants without AD. The results suggest that sCD14 in BM may be involved in atopic manifestations in early infancy.
Collapse
|
45
|
Hu P, Zhao F, Zhu W, Wang J. Effects of early-life lactoferrin intervention on growth performance, small intestinal function and gut microbiota in suckling piglets. Food Funct 2019; 10:5361-5373. [PMID: 31393487 DOI: 10.1039/c9fo00676a] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The early postnatal stage is a critical period for suckling animals in developing intestinal function and stabilizing gut microbiota. Lactoferrin (LF) plays a critical role in promoting gut development and regulating gut microbiota. This study investigates the impact of early-life lactoferrin (LF) intervention on the growth performance, small intestinal function and gut microbiota in suckling piglets. Sixty suckling piglets (1.51 ± 0.05 kg) obtained from six sows (10 piglets per litter) were assigned to a control (CON) group and an LF group in each litter, which were sow-fed. Piglets in the LF group were orally administered 8-12 mL LF solution (0.5 g per kg body weight per day) for a week, and piglets in the CON group were orally administered the same dose of physiological saline. Six piglets (n = 6) from each group were euthanized on days 8 and 21. The early-life LF intervention increased growth performance, with higher villi height of the jejunum and greater disaccharidase activity of the jejunum and ileum (P < 0.05). Diarrhoea incidence decreased in the LF group from day 1 to day 7 (P < 0.05). Urinary lactulose-mannitol ratios decreased in the LF group, whereas the gene and protein expressions of jejunal occludin increased in the LF group on day 8 and day 21, and higher gene and protein levels of ileal occludin were observed on day 8 (P < 0.05). Additionally, the LF piglets had lower concentrations of IL-1β and TNF-α, and higher concentration of IL-10 in the jejunum (P < 0.05). For the ileum, higher concentration of IL-10 and lower concentration of TNF-α were observed in the LF group (P < 0.05). LF piglets had a greater abundance of Lactobacillus and lower abundance of Veillonella and Escherichia-Shigella in the jejunum on day 8 (P < 0.05). In the ileum, the abundance of Actinobacillus was decreased in the LF piglets on day 8 and day 21 (P < 0.05). The early-life LF intervention enhanced the growth performance and decreased diarrhoea incidence in the suckling piglets by promoting the development of intestinal function and changing the microbiota in the small intestine.
Collapse
Affiliation(s)
- Ping Hu
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu Province 210095, P. R. China. and Laboratory of Gastrointestinal Microbiology, Xiaolingwei Street, Weigang 1, Nanjing 210095, China
| | - Fangzhou Zhao
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu Province 210095, P. R. China. and Laboratory of Gastrointestinal Microbiology, Xiaolingwei Street, Weigang 1, Nanjing 210095, China
| | - Weiyun Zhu
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu Province 210095, P. R. China. and Laboratory of Gastrointestinal Microbiology, Xiaolingwei Street, Weigang 1, Nanjing 210095, China
| | - Jing Wang
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu Province 210095, P. R. China. and Laboratory of Gastrointestinal Microbiology, Xiaolingwei Street, Weigang 1, Nanjing 210095, China
| |
Collapse
|
46
|
Boix-Amorós A, Collado MC, Van't Land B, Calvert A, Le Doare K, Garssen J, Hanna H, Khaleva E, Peroni DG, Geddes DT, Kozyrskyj AL, Warner JO, Munblit D. Reviewing the evidence on breast milk composition and immunological outcomes. Nutr Rev 2019; 77:541-556. [PMID: 31111150 DOI: 10.1093/nutrit/nuz019] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
A large number of biologically active components have been found in human milk (HM), and in both human and animal models, studies have provided some evidence suggesting that HM composition can be altered by maternal exposures, subsequently influencing health outcomes for the breastfed child. Evidence varies from the research studies on whether breastfeeding protects the offspring from noncommunicable diseases, including those associated with immunological dysfunction. It has been hypothesized that the conflicting evidence results from HM composition variations, which contain many immune active molecules, oligosaccharides, lactoferrin, and lysozyme in differing concentrations, along with a diverse microbiome. Determining the components that influence infant health outcomes in terms of both short- and long-term sequelae is complicated by a lack of understanding of the environmental factors that modify HM constituents and thereby offspring outcomes. Variations in HM immune and microbial composition (and the differing infantile responses) may in part explain the controversies that are evidenced in studies that aim to evaluate the prevalence of allergy by prolonged and exclusive breastfeeding. HM is a "mixture" of immune active factors, oligosaccharides, and microbes, which all may influence early immunological outcomes. This comprehensive review provides an in-depth overview of existing evidence on the studied relationships between maternal exposures, HM composition, vaccine responses, and immunological outcomes.
Collapse
Affiliation(s)
- Alba Boix-Amorós
- Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
| | - Belinda Van't Land
- Department of Immunology, Danone Nutricia Research, Utrecht, the Netherlands
- Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Anna Calvert
- Paediatric Infectious Diseases Research Group, St George's University of London, London, United Kingdom
| | - Kirsty Le Doare
- Paediatric Infectious Diseases Research Group, St George's University of London, London, United Kingdom
- Imperial College London, London, United Kingdom
- Public Health England, Porton Down, United Kingdom, and the MRC Unit, Fajara, Gambia
| | - Johan Garssen
- Department of Immunology, Danone Nutricia Research, Utrecht, the Netherlands
- Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | | | - Ekaterina Khaleva
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- University of Southampton, Southampton, UK
| | - Diego G Peroni
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- Department of Clinical and Experimental Medicine, Section of Paediatrics, University of Pisa, Pisa, Italy
| | - Donna T Geddes
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- School of Molecular Sciences, the University of Western Australia, Perth, Australia
| | - Anita L Kozyrskyj
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- Department of Pediatrics, Department of Obstetrics & Gynecology, Faculty of Medicine & Dentistry, School of Public Health, University of Alberta, Alberta, Canada
| | - John O Warner
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- Imperial College London, London, United Kingdom
- National Institute for Health Research, Collaboration for Leadership in Applied Health Research and Care for NW London, London, United Kingdom
| | - Daniel Munblit
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- Imperial College London, London, United Kingdom
- Department of Pediatrics, Sechenov University, Moscow, Russia, and the Solov'ev Research and Clinical Center for Neuropsychiatry, Moscow, Russia
| |
Collapse
|
47
|
Abstract
Supplementation of probiotics to very low birth weight (VLBW) infants has been extensively studied, with multiple meta-analyses reporting probiotics decrease the risk of necrotizing enterocolitis (NEC) and death. Despite availability of this evidence, the decision to initiate routine probiotic supplementation to preterm infants continues to be a complex one. There are uncertainties regarding the use of probiotics, including selecting the appropriate product, dose and target population. Additionally, availability of specific probiotic products and regulatory oversight varies by country, raising concerns regarding the safety and efficacy of specific probiotic products. In this review, we summarize the latest evidence on probiotic use in preterm infants and discuss considerations that may help guide clinicians who are considering routine probiotic supplementation.
Collapse
|
48
|
Longitudinal Analysis of Macronutrient Composition in Preterm and Term Human Milk: A Prospective Cohort Study. Nutrients 2019; 11:nu11071525. [PMID: 31277502 PMCID: PMC6683284 DOI: 10.3390/nu11071525] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/14/2019] [Accepted: 07/01/2019] [Indexed: 12/22/2022] Open
Abstract
Background: Mother’s own milk is the optimal source of nutrients and provides numerous health advantages for mothers and infants. As they have supplementary nutritional needs, very preterm infants may require fortification of human milk (HM). Addressing HM composition and variations is essential to optimize HM fortification strategies for these vulnerable infants. Aims: To analyze and compare macronutrient composition in HM of mothers lactating very preterm (PT) (28 0/7 to 32 6/7 weeks of gestational age, GA) and term (T) infants (37 0/7 to 41 6/7 weeks of GA) over time, both at similar postnatal and postmenstrual ages, and to investigate other potential factors of variations. Methods: Milk samples from 27 mothers of the PT infants and 34 mothers of the T infants were collected longitudinally at 12 points in time during four months for the PT HM and eight points in time during two months for the T HM. Macronutrient composition (proteins, fat, and lactose) and energy were measured using a mid-infrared milk analyzer, corrected by bicinchoninic acid (BCA) assay for total protein content. Results: Analysis of 500 HM samples revealed large inter- and intra-subject variations in both groups. Proteins decreased from birth to four months in the PT and the T HM without significant differences at any postnatal time point, while it was lower around term equivalent age in PT HM. Lactose content remained stable and comparable over time. The PT HM contained significantly more fat and tended to be more caloric in the first two weeks of lactation, while the T HM revealed higher fat and higher energy content later during lactation (three to eight weeks). In both groups, male gender was associated with more fat and energy content. The gender association was stronger in the PT group, and it remained significant after adjustments. Conclusion: Longitudinal measurements of macronutrients compositions of the PT and the T HM showed only small differences at similar postnatal stages in our population. However, numerous differences exist at similar postmenstrual ages. Male gender seems to be associated with a higher content in fat, especially in the PT HM. This study provides original information on macronutrient composition and variations of HM, which is important to consider for the optimization of nutrition and growth of PT infants.
Collapse
|
49
|
Khandelwal P, Andersen H, Romick-Rosendale L, Taggart CB, Watanabe M, Lane A, Dandoy CE, Lake KE, Litts BA, Morrow AL, Lee ML, Haslam DB, Davies SM. A Pilot Study of Human Milk to Reduce Intestinal Inflammation After Bone Marrow Transplant. Breastfeed Med 2019; 14:193-202. [PMID: 30916575 DOI: 10.1089/bfm.2018.0199] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Human milk administration in the early peritransplant period would lower intestinal inflammation after bone marrow transplant (BMT). MATERIALS AND METHODS Children 0-5 years undergoing BMT received either a ready-to-feed human milk preparation designed for these children (Prolacta Bioscience, Duarte, CA) or standard formula. Babies breastfeeding at the time of BMT were also enrolled on the human milk arm. Human milk was administered from day -3 until day +14 after BMT. Metagenomic shotgun sequencing and metabolomics of stool, plasma cytokines, and regenerating islet-derived 3α (REG3α) levels were measured at enrollment and day +14. Human leukocyte antigen-DR isotype (HLA-DR), CD38, and CD69 expression on T cells were evaluated at day +21. RESULTS Forty-six children were enrolled, 32 received human milk (donor milk n = 23, breastfeeding babies n = 9), and 14 were controls who received standard feeds supervised by a BMT dietician. Twenty-four patients received at least 60% of goal human milk and were evaluable. Plasma interleukin (IL)-8 (p = 0.04), IL-10 (p = 0.02), and REG3α (p = 0.03) were decreased in the human milk cohort. Peripheral blood CD69+ CD8+ T cells were higher in controls (p = 0.01). Species abundance of Adenovirus (p = 0.00034), Escherichia coli (p = 0.0017), Cryptosporidium parvum (p = 0.0006), Dialister invisus (p = 0.01), and Pseudomonas aeruginosa (p = 0.05) from stool was higher in controls. Stool alanine, tyrosine, methionine, and the ratio of fecal alanine to choline and phosphocholine were higher in controls (p < 0.05). No difference was observed in stool propionate and butyrate levels as measures of short-chain fatty acids between the two cohorts. CONCLUSIONS Administration of human milk resulted in decreased markers of intestinal inflammation and could be a valuable adjunct for patients after BMT.
Collapse
Affiliation(s)
- Pooja Khandelwal
- 1 Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Heidi Andersen
- 2 Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | | | - Cynthia B Taggart
- 4 Department of Nutrition Therapy, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Miki Watanabe
- 3 Divison of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Adam Lane
- 1 Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Christopher E Dandoy
- 1 Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kelly E Lake
- 1 Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Bridget A Litts
- 1 Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Ardythe L Morrow
- 5 Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | | | - David B Haslam
- 2 Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Stella M Davies
- 1 Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
50
|
A new dilution-enrichment sample preparation strategy for expanded metabolome monitoring of human breast milk that overcomes the simultaneous presence of low- and high-abundance lipid species. Food Chem 2019; 288:154-161. [PMID: 30902276 DOI: 10.1016/j.foodchem.2019.03.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/01/2019] [Accepted: 03/03/2019] [Indexed: 11/23/2022]
Abstract
The complex nature of human breast milk (HBM) makes samples difficult to analyze, requiring several extraction techniques and analytical platforms to obtain high metabolome coverage. In this work, we combined liquid-liquid extraction (LLE) and solid-phase extraction (SPE) techniques to prepare HBM samples to overcome the challenge of low- and high-abundance lipid species, enabling the semiquantitative analysis of total HBM lipids in one liquid chromatography-mass spectrometry (LC-MS) run. A nonorganic fraction obtained during the LLE step was used to analyze small polar metabolites. This analytical approach allows extensive metabolome coverage, especially for low-abundance glycerophospholipids and sphingolipids. The method was applied to monitor short-term metabolome changes in HBM composition within individual mothers and the results showed variable metabolite composition patterns. Simultaneous detection of high-abundance glycerolipids and low-abundance but not less significant phospholipids in one LC-MS run saves time, decreases cost, and enables comprehensive insight into the dynamics of HBM composition.
Collapse
|