1
|
Thomas KL, Wahlquist AE, James D, Clark WA, Wagner CL. Effects of Maternal Pre-Pregnancy BMI on Preterm Infant Microbiome and Fecal Fermentation Profile-A Preliminary Cohort Study. Nutrients 2025; 17:987. [PMID: 40290029 PMCID: PMC11944810 DOI: 10.3390/nu17060987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/05/2025] [Accepted: 03/05/2025] [Indexed: 04/30/2025] Open
Abstract
Objective: This feasibility, proof-of-concept study aimed to assess the impact of maternal pre-pregnancy body mass index (BMI) on preterm infant fecal fermentation and microbiome. Study Design: An infant cohort study (n = 54) in the NICU at MUSC from June 2021 to September 2022 was grouped according to maternal pre-pregnancy BMI-normal weight (<25 kg/m2), overweight (25-29.9 kg/m2), and obese (≥30 kg/m2). All fecal samples were subjected to 16s rRNA isolation and analysis, as well as short chain fatty acid (SCFA) extraction and analysis. Results: Preterm infants born to overweight and obese mothers did not have differences in microbial diversities but did have different bacterial taxonomic composition and lower relative abundance levels of taxa than those born to normal-weight mothers. While controlling for covariates, we found SCFA propionic acid to be higher and more significant in infant stools born to mothers with a higher pre-pregnancy BMI. Conclusions: This is a novel study investigating the microbiome and SCFA in premature infants while considering maternal pre-pregnancy BMI. This study adds to the current literature, in that the preterm infant gut is generally lower in microbial diversity which can impact infant health. Thus, it is important to understand the mechanisms necessary to modulate the microbiome of preterm infants to improve their health outcomes.
Collapse
Affiliation(s)
- Kristy L. Thomas
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (K.L.T.); (W.A.C.)
- Department of Rehabilitative Sciences, College of Health Sciences, East Tennessee State University, Johnson City, TN 37614, USA
| | - Amy E. Wahlquist
- Center for Rural Health and Research, Department of Biostatistics and Epidemiology, College of Public Health, East Tennessee State University, Johnson City, TN 37614, USA
| | - Dalton James
- Department of Biological Sciences, College of Arts and Sciences, East Tennessee State University, Johnson City, TN 37614, USA
| | - William Andrew Clark
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (K.L.T.); (W.A.C.)
- Department of Rehabilitative Sciences, College of Health Sciences, East Tennessee State University, Johnson City, TN 37614, USA
| | - Carol L. Wagner
- Division of Neonatology, Department of Pediatrics, Shawn Jenkins Children’s Hospital, Medical University of South Carolina, 10 McClennan Banks Drive, MSC 915, Charleston, SC 29425, USA
| |
Collapse
|
2
|
May MF, McKinney ML, Cestare D, Hussey A, Zevallos AB, Garber S, Posencheg MA, Puopolo KM, Mukhopadhyay S. Improving Time to First Feeding for Preterm Infants: A Quality Improvement Approach. Pediatr Qual Saf 2025; 10:e798. [PMID: 39980492 PMCID: PMC11841844 DOI: 10.1097/pq9.0000000000000798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 02/04/2025] [Indexed: 02/22/2025] Open
Abstract
Introduction Early initiation of enteral feeding is reported to reduce sepsis and mortality in preterm infants. Less than half of stable infants born <35 weeks gestational age with birth weight <2,000 g are fed within 24 hours of birth at our center. Methods The Specific, measureable, achievable, relevant, timely aim of this quality improvement project was to increase the initiation of enteral feeding within 24 hours of birth from 49% (baseline) to 75% among infants born <35 weeks gestational age with birth weight <2,000 g between November 2022 and December 2023. We identified the unavailability of a mother's own milk as a major barrier to early feeding initiation. Project interventions included antenatal lactation consultation, patient-dedicated breast pumps, standardized feeding orders, and multidisciplinary education. The outcome measure was the time to first enteral feeding, and the balancing measures were the diagnosis of necrotizing enterocolitis (NEC) and the use of formula for first feeding. Results The proportion of infants with feeding initiated within 24 hours of birth increased from 49% to 80% during the project period. The incidence of NEC was unchanged (1.9% before and during the project period). Both before and during the project, feedings were most frequently initiated with pasteurized donor human milk (49.7% versus 58.7%), followed by mother's own milk (37.8% versus 35.6%) and formula (12.5% versus 5.8%). Conclusions This quality improvement project increased the proportion of eligible infants fed within 24 hours of birth without a change in the incidence of NEC or an increase in formula use as first feeding.
Collapse
Affiliation(s)
- Molly F. May
- From the Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pa
- CHOP Newborn Care at Pennsylvania Hospital, Philadelphia, Pa
- University of Pittsburgh School of Nursing, Pittsburgh, PA
| | | | - Danielle Cestare
- From the Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pa
| | - Alicia Hussey
- CHOP Newborn Care at Pennsylvania Hospital, Philadelphia, Pa
| | - Alvaro Barboza Zevallos
- From the Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pa
- Clinical Futures, Children’s Hospital of Philadelphia Research Institute, Philadelphia, Pa
| | - Samuel Garber
- From the Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pa
- CHOP Newborn Care at Pennsylvania Hospital, Philadelphia, Pa
| | - Michael A. Posencheg
- From the Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pa
- Clinical Futures, Children’s Hospital of Philadelphia Research Institute, Philadelphia, Pa
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Karen M. Puopolo
- From the Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pa
- CHOP Newborn Care at Pennsylvania Hospital, Philadelphia, Pa
- Clinical Futures, Children’s Hospital of Philadelphia Research Institute, Philadelphia, Pa
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Sagori Mukhopadhyay
- From the Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pa
- CHOP Newborn Care at Pennsylvania Hospital, Philadelphia, Pa
- Clinical Futures, Children’s Hospital of Philadelphia Research Institute, Philadelphia, Pa
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| |
Collapse
|
3
|
Lawrence SM, Wynn JL, Gordon SM. Neonatal bacteremia and sepsis. REMINGTON AND KLEIN'S INFECTIOUS DISEASES OF THE FETUS AND NEWBORN INFANT 2025:183-232.e25. [DOI: 10.1016/b978-0-323-79525-8.00015-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
4
|
Chen J, van Wesemael AJ, Denswil NP, Niemarkt HJ, van Goudoever JB, Muncan V, de Meij TG, van den Akker CH. Impact of mother's own milk vs. donor human milk on gut microbiota colonization in preterm infants: a systematic review. MICROBIOME RESEARCH REPORTS 2024; 4:8. [PMID: 40207271 PMCID: PMC11977380 DOI: 10.20517/mrr.2024.44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/31/2025] [Accepted: 11/11/2024] [Indexed: 04/11/2025]
Abstract
Background: Nutritional intake in preterm infants is associated with short- and long-term outcomes. The favorable outcomes of preterm infants who predominantly receive their mother's own milk (MOM) are thought to be mediated partly through beneficial effects on the gut microbiome. When MOM is not available, donor human milk (DHM) is recommended as the best alternative. However, DHM is less effective in preventing adverse outcomes, which may be explained by compositional differences between MOM and DHM, resulting in different microbiome development. This systematic review focuses on the effects of predominant DHM vs. MOM feeding on the gut microbiota composition in preterm infants. Methods: A comprehensive search was conducted across MEDLINE, Embase, and Cochrane databases. Eight out of the 717 publications identified were included. Data on gut microbiota composition, alpha diversity, and taxonomic differences between DHM- and MOM-fed preterm infants were extracted and analyzed. Results: The microbiome composition was distinct between the two feeding groups. Alpha diversity measures were lower in DHM-fed infants, particularly when preterm formula (PF) was also provided. DHM-fed infants showed higher abundances of Staphylococcaceae and Clostridiaceae, and lower abundances of Bacteroidetes and Bifidobacterium. Conclusion: The observed gut microbiome differences in DHM-fed preterm infants have previously been linked to adverse health outcomes. This underlines the importance of increasing the awareness of MOM intake in preterm infants. Further studies should explore the mechanisms through which human milk affects health outcomes.
Collapse
Affiliation(s)
- Jing Chen
- Department of Neonatology, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam 1105 AZ, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam UMC, Amsterdam 1105 AZ, the Netherlands
- Authors contributed equally
| | - Aranka J. van Wesemael
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam 1105 AZ, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam UMC, Amsterdam 1105 AZ, the Netherlands
- Authors contributed equally
| | - Nerissa P. Denswil
- Medical Library, Amsterdam UMC, location University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Hendrik J. Niemarkt
- Department of Neonatology, Maxima Medical Centre, Veldhoven 5504 DB, the Netherlands
- Department of Electrical Engineering, Technical University Eindhoven, Eindhoven 5612 AZ, the Netherlands
| | - Johannes B. van Goudoever
- Department of Neonatology, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam 1105 AZ, the Netherlands
| | - Vanesa Muncan
- Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam UMC, Amsterdam 1105 AZ, the Netherlands
| | - Tim G.J. de Meij
- Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam UMC, Amsterdam 1105 AZ, the Netherlands
- Department of Pediatric Gastroenterology, Emma Children’s Hospital, Amsterdam UMC, location University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
- Authors contributed equally
| | - Chris H.P. van den Akker
- Department of Neonatology, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam 1105 AZ, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam UMC, Amsterdam 1105 AZ, the Netherlands
- Authors contributed equally
| |
Collapse
|
5
|
Hutchinson R, Wade W, Millar M, Ansbro K, Stacey F, Costeloe K, Fleming P. Changes in the intestinal microbiome of the preterm baby associated with stopping non-invasive pressure support: a prospective cohort study. BMJ Paediatr Open 2024; 8:e002675. [PMID: 39362793 PMCID: PMC11459353 DOI: 10.1136/bmjpo-2024-002675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 09/09/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Intestinal dysbiosis is implicated in the pathogenesis of necrotising enterocolitis and late-onset sepsis in preterm babies. The provision of non-invasive positive pressure ventilation is a common clinical intervention in preterm babies, and may be hypothesised to adversely affect intestinal bacterial growth, through increased aerophagia and induction of a hyperoxic intestinal environment; however this relationship has not been previously well characterised. METHODOLOGY In this prospectively recruited cohort study, high-throughput 16S rRNA gene sequencing was combined with contemporaneous clinical data collection, to assess within-subject changes in microbiome development around the time of transitioning from non-invasive positive pressure respiratory support to unsupported spontaneous breathing. RESULTS In a group of 14 preterm infants, bacterial diversity was seen to increase by 0.34 units/week (inverse Simpson index) at the point of transitioning off non-invasive positive pressure respiratory support. Correspondingly, a significant increase in anaerobic genera (Bifidobacteria spp, Veillonella spp), and a non-significant fall in Enterobacteriaceae was also seen at this time. CONCLUSIONS Provision of non-invasive positive pressure ventilation is associated with suppression of both diversity accrual and obligate anaerobic growth in the preterm intestine. This has clinical implications in view of the widespread use of non-invasive positive pressure ventilation in preterm neonatal care (and wider adult use), and demonstrates the need for potential strategies (eg, probiotic support; reduced aerophagia) to support the development of a healthy gut microbiome during this time.
Collapse
Affiliation(s)
- Richard Hutchinson
- Queen Mary University of London, London, UK
- Department of Neonatology, Homerton University Hospital NHS Foundation Trust, London, UK
| | - William Wade
- King's College London, London, UK
- The Forsyth Institute, Cambridge, Massachusetts, USA
| | | | | | - Fiona Stacey
- Department of Neonatology, Homerton University Hospital NHS Foundation Trust, London, UK
| | - Kate Costeloe
- Department of Neonatology, Homerton University Hospital NHS Foundation Trust, London, UK
- Genomics and Child Health, Queen Mary University of London Barts and The London School of Medicine and Dentistry, London, UK
| | - Paul Fleming
- Department of Neonatology, Homerton University Hospital NHS Foundation Trust, London, UK
- Genomics and Child Health, Queen Mary University of London Barts and The London School of Medicine and Dentistry, London, UK
| |
Collapse
|
6
|
Waller ME, Eichhorn CJ, Gutierrez A, Baatz JE, Wagner CL, Chetta KE, Engevik MA. Analyzing the Responses of Enteric Bacteria to Neonatal Intensive Care Supplements. Int J Microbiol 2024; 2024:3840327. [PMID: 39220439 PMCID: PMC11364479 DOI: 10.1155/2024/3840327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
In the neonatal intensive care unit, adequate nutrition requires various enteral products, including human milk and formula. Human milk is typically fortified to meet increased calorie goals, and infants commonly receive vitamin mixes, iron supplements, and less frequently, thickening agents. We examined the growth of 16 commensal microbes and 10 pathobionts found in the premature infant gut and found that formula, freshly pasteurized milk, and donated banked milk generally increased bacterial growth. Fortification of human milk significantly elevated the growth of all microbes. Supplementation with thickeners or NaCl in general did not stimulate additional growth. Vitamin mix promoted the growth of several commensals, while iron promoted growth of pathobionts. These data indicate that pathobionts in the preterm gut have significant growth advantage with preterm formula, fortified donor milk, and supplemented iron and suggest that the choice of milk and supplements may impact the infant gut microbiota.
Collapse
Affiliation(s)
- Megan E. Waller
- Department of Regenerative Medicine and Cell BiologyMedical University of South Carolina, Charleston, USA
| | - Caroline J. Eichhorn
- Department of Regenerative Medicine and Cell BiologyMedical University of South Carolina, Charleston, USA
| | - Alyssa Gutierrez
- Department of Regenerative Medicine and Cell BiologyMedical University of South Carolina, Charleston, USA
| | - John E. Baatz
- Department of PediatricsC.P. Darby Children's Research InstituteMedical University of South Carolina, Charleston, USA
- Department of PediatricsDivision of Neonatal-Perinatal MedicineMedical University of South CarolinaShawn Jenkins Children's Hospital, 10 McClennan Banks Drive, MSC 915, Charleston, SC 29425, USA
| | - Carol L. Wagner
- Department of PediatricsC.P. Darby Children's Research InstituteMedical University of South Carolina, Charleston, USA
- Department of PediatricsDivision of Neonatal-Perinatal MedicineMedical University of South CarolinaShawn Jenkins Children's Hospital, 10 McClennan Banks Drive, MSC 915, Charleston, SC 29425, USA
| | - Katherine E. Chetta
- Department of PediatricsC.P. Darby Children's Research InstituteMedical University of South Carolina, Charleston, USA
- Department of PediatricsDivision of Neonatal-Perinatal MedicineMedical University of South CarolinaShawn Jenkins Children's Hospital, 10 McClennan Banks Drive, MSC 915, Charleston, SC 29425, USA
| | - Melinda A. Engevik
- Department of Regenerative Medicine and Cell BiologyMedical University of South Carolina, Charleston, USA
- Department of Microbiology and ImmunologyMedical University of South Carolina, Charleston, USA
| |
Collapse
|
7
|
Pütz E, Ascherl R, Wendt T, Thome UH, Gebauer C, Genuneit J, Siziba LP. The association of different types of human milk with bronchopulmonary dysplasia in preterm infants. Front Nutr 2024; 11:1408033. [PMID: 39171103 PMCID: PMC11337300 DOI: 10.3389/fnut.2024.1408033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/15/2024] [Indexed: 08/23/2024] Open
Abstract
Objective To evaluate the association between different types of human milk feeds and bronchopulmonary dysplasia (BPD) in preterm infants. Methods Data on dispensed mother's own milk (MOM) and donor human milk (DHM) from Leipzig Milk Bank for hospitalized infants with a gestational age (GA) ≤32 weeks observed from birth to 36 weeks' postmenstrual age or prior discharge were used. BPD was assessed based on documented International Statistical Classification of Diseases and Related Health Problems (ICD) diagnosis and on electronic hospital records (EHR) of data on ventilation and oxygen supplementation. Associations of dispensed milk feed variations with BPD were investigated using logistic regressions in crude and adjusted models. Results 866 infants were included with a BPD prevalence of 15.4% (EHR) and 23.2% (ICD). The mean GA was 29.1 weeks. The majority (84.4%, n = 746) of infants were nurtured with a mix of MOM, DHM supplemented by formula or parenteral (other) nutrition during hospitalization. For which, MOM comprised the highest median [Q1-Q3] percentage proportion (53[31-81] %) of this mix. Exclusive fresh milk and exclusive MOM feeds were dispensed on a mean of 40 and 34% patient-days, respectively. Statistically significant associations with lower BPD incidence were only observed for 70-80% MOM vs. DHM, and 60% fresh vs. frozen milk, in crude and adjusted models. Conclusion Our findings suggest a protective association of MOM and fresh milk with lower odds of BPD, which may be dependent on the proportion of MOM or fresh milk administered. These results highlight the importance of MOM as an ideal source of nutrition during early infancy.
Collapse
Affiliation(s)
- Elisabeth Pütz
- Pediatric Epidemiology, Department of Pediatrics, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Rudolf Ascherl
- Division of Neonatology, Department of Pediatrics, University of Leipzig Medical Center, Leipzig, Germany
| | - Thomas Wendt
- Data Integration Center, University of Leipzig Medical Center, Leipzig, Germany
| | - Ulrich H. Thome
- Division of Neonatology, Department of Pediatrics, University of Leipzig Medical Center, Leipzig, Germany
| | - Corinna Gebauer
- Division of Neonatology, Department of Pediatrics, University of Leipzig Medical Center, Leipzig, Germany
| | - Jon Genuneit
- Pediatric Epidemiology, Department of Pediatrics, Medical Faculty, Leipzig University, Leipzig, Germany
- German Center for Child and Youth Health (DZKJ), Leipzig, Germany
| | - Linda P. Siziba
- Pediatric Epidemiology, Department of Pediatrics, Medical Faculty, Leipzig University, Leipzig, Germany
| |
Collapse
|
8
|
van den Elsen LJW, Rekima A, Lynn MA, Isnard C, Machado S, Divakara N, Patalwala D, Middleton A, Stevens N, Servant F, Burcelin R, Lynn DJ, Verhasselt V. Diet at birth is critical for healthy growth, independent of effects on the gut microbiota. MICROBIOME 2024; 12:139. [PMID: 39068488 PMCID: PMC11282663 DOI: 10.1186/s40168-024-01852-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 05/30/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Colostrum is the first milk for a newborn. Its high content in microbiota shaping compounds and its intake at the time of gut microbiota seeding suggests colostrum may be critical in the establishment of a healthy microbiota. There is also accumulating evidence on the importance of the gut microbiota for healthy growth. Here, we aimed to investigate the contribution of colostrum, and colostrum-induced microbiota to growth promotion. Addressing this question is highly significant because (1) globally, less than half of the newborns are fully colostrum fed (2) the evidence for the importance of the microbiota for the prevention of undernutrition has only been demonstrated in juvenile or adult pre-clinical models while stunting already starts before weaning. RESULTS To address the importance of diet at birth in growth failure, we developed a unique mouse model in which neonates are breastfed by mothers at an advanced stage of lactation who no longer provide colostrum. Feeding newborn mice with mature milk instead of colostrum resulted in significant growth retardation associated with the biological features of chronic undernutrition, such as low leptin levels, dyslipidemia, systemic inflammation, and growth hormone resistance. We next investigated the role of colostrum in microbiota shaping. At the end of the lactation period, we found a major difference in gut microbiota alpha diversity, beta diversity, and taxa distribution in control and colostrum-deprived mice. To determine the causal relationship between changes in microbiota and growth trajectories, we repeated our experiment in germ-free mice. The beneficial effect of colostrum on growth remained in the absence of microbiota. CONCLUSION Our data suggest that colostrum may play an important role in the prevention of growth failure. They highlight that the interplay between neonatal gut microbiome assembly and diet may not be as crucial for growth control in the developing newborn as described in young adults. This opens a paradigm shift that will foster research for colostrum's bioactives that may exert a similar effect to microbiota-derived ligands in promoting growth and lead to new avenues of translational research for newborn-tailored prevention of stunting. Video Abstract.
Collapse
Affiliation(s)
- Lieke J W van den Elsen
- Larsson-Rosenquist Centre for Immunology and Breastfeeding, School of Medicine, The University of Western Australia, Perth, WA, Australia.
- Telethon Kids Institute, Perth, WA, Australia.
| | - Akila Rekima
- Larsson-Rosenquist Centre for Immunology and Breastfeeding, School of Medicine, The University of Western Australia, Perth, WA, Australia
- Telethon Kids Institute, Perth, WA, Australia
| | - Miriam A Lynn
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Flinders Health and Medical Research Institute, Flinders University, Adelaide, SA, Australia
| | | | - Savannah Machado
- Larsson-Rosenquist Centre for Immunology and Breastfeeding, School of Medicine, The University of Western Australia, Perth, WA, Australia
- Telethon Kids Institute, Perth, WA, Australia
| | - Nivedithaa Divakara
- Larsson-Rosenquist Centre for Immunology and Breastfeeding, School of Medicine, The University of Western Australia, Perth, WA, Australia
- Telethon Kids Institute, Perth, WA, Australia
| | - Diana Patalwala
- National Imaging Facility, Centre for Microscopy Characterisation and Analysis, University of Western Australia, Perth, WA, Australia
| | - Alana Middleton
- Larsson-Rosenquist Centre for Immunology and Breastfeeding, School of Medicine, The University of Western Australia, Perth, WA, Australia
| | - Natalie Stevens
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Flinders Health and Medical Research Institute, Flinders University, Adelaide, SA, Australia
| | | | | | - David J Lynn
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Flinders Health and Medical Research Institute, Flinders University, Adelaide, SA, Australia
| | - Valerie Verhasselt
- Larsson-Rosenquist Centre for Immunology and Breastfeeding, School of Medicine, The University of Western Australia, Perth, WA, Australia.
- Telethon Kids Institute, Perth, WA, Australia.
| |
Collapse
|
9
|
Strle A, Reyes SM, Schmidt M, Lynch MF. Human Milk-Derived Fortifier to Reduce Hospital-Acquired Malnutrition in Uncomplicated Gastroschisis: A Case Report. AJP Rep 2024; 14:e275-e280. [PMID: 39717541 PMCID: PMC11666322 DOI: 10.1055/a-2490-3521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/22/2024] [Indexed: 12/25/2024] Open
Abstract
Gastroschisis is one of the most common congenital gastrointestinal disorders, occurring in about one in 1,953 infants born each year in the United States. Infants with gastroschisis rely on total parenteral nutrition (TPN) preoperatively, and due to intestinal function and dysmotility issues, continue to face feeding challenges postclosure, including feeding intolerance and increased risk of necrotizing enterocolitis (NEC). Postclosure, human milk-feeding is preferred over infant formula because of its associated reduced risk of feeding intolerance and NEC. However, unfortified human milk often falls short of meeting the increased metabolic demands of these postsurgical infants in the first few weeks of life, leading to hospital-acquired malnutrition (undernutrition) as TPN is weaned. We hypothesized that fortifying maternal milk with human milk-based fortifiers would mitigate the risk of hospital-acquired malnutrition while providing the tolerance benefits of an exclusive human milk diet, specifically by meeting the increased energy and protein demands of the immediate postsurgical infant as parenteral nutrition is weaned. The case report describes our unit's use of a human milk-based fortifier in an infant with uncomplicated gastroschisis and its positive effect on the patient's growth. Further research is warranted to assess the use of human milk-derived fortifiers to prevent hospital-acquired malnutrition after gastrointestinal surgery.
Collapse
Affiliation(s)
- Anna Strle
- Neonatal Intensive Care Unit, Texas Health Harris Methodist Hospital Fort Worth, Fort Worth, Texas
| | | | - Megan Schmidt
- Neonatal Intensive Care Unit, Texas Health Harris Methodist Hospital Fort Worth, Fort Worth, Texas
| | - Mary Frances Lynch
- Neonatal Intensive Care Unit, Texas Health Harris Methodist Hospital Fort Worth, Fort Worth, Texas
| |
Collapse
|
10
|
Younge N. Influence of infant microbiome on health and development. Clin Exp Pediatr 2024; 67:224-231. [PMID: 37605538 PMCID: PMC11065641 DOI: 10.3345/cep.2023.00598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/08/2023] [Accepted: 07/19/2023] [Indexed: 08/23/2023] Open
Abstract
The microbiome is a complex ecosystem comprising microbes, their genomes, and the surrounding environment. The microbiome plays a critical role in early human development, including maturation of the host immune system and gastrointestinal tract. Multiple factors, including diet, anti-biotic use, and other environmental exposures, influence the establishment of the microbiome during infancy. Numerous studies have identified associations between the microbiome and neonatal diseases, including necrotizing enterocolitis, sepsis, and malnutrition. Furthermore, there is compelling evidence that perturbation of the microbiome in early life can have lasting developmental effects that increase an individual's risk for immune and metabolic diseases in later life. Supplementation of the microbiome with probiotics reduces the risk of necrotizing enterocolitis and sepsis in at-risk infants. This review focuses on the structure and function of the infant microbiome, the environmental and clinical factors that influence its assembly, and its impact on infant health and development.
Collapse
|
11
|
Adeniyi-Ipadeola GO, Hoffman KL, Yang H, Javornik Cregeen SJ, Preidis GA, Ramani S, Hair AB. Human milk cream alters intestinal microbiome of preterm infants: a prospective cohort study. Pediatr Res 2024; 95:1564-1571. [PMID: 38228744 DOI: 10.1038/s41390-023-02948-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 11/09/2023] [Accepted: 11/18/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND In very low birth weight (VLBW) infants, human milk cream added to standard human milk fortification is used to improve growth. This study aimed to evaluate the impact of cream supplement on the intestinal microbiome of VLBW infants. METHODS Whole genome shotgun sequencing was performed on stool (n = 57) collected from a cohort of 23 infants weighing 500-1250 grams (control = 12, cream = 11). Both groups received an exclusive human milk diet (mother's own milk, donor human milk, and donor human milk-derived fortifier) with the cream group receiving an additional 2 kcal/oz cream at 100 mL/kg/day of fortified feeds and then 4 kcal/oz if poor growth. RESULTS While there were no significant differences in alpha diversity, infants receiving cream significantly differed from infants in the control group in beta diversity. Cream group samples had significantly higher prevalence of Proteobacteria and significantly lower Firmicutes compared to control group. Klebsiella species dominated the microbiota of cream-exposed infants, along with bacterial pathways involved in lipid metabolism and metabolism of cofactors and amino acids. CONCLUSIONS Cream supplementation significantly altered composition of the intestinal microbiome of VLBW infants to favor increased prevalence of Proteobacteria and functional gene content associated with these bacteria. IMPACT We report changes to the intestinal microbiome associated with administration of human milk cream; a novel supplement used to improve growth rates of preterm very low birth weight infants. Since little is known about the impact of cream on intestinal microbiota composition of very low birth weight infants, our study provides valuable insight on the effects of diet on the microbiome of this population. Dietary supplements administered to preterm infants in neonatal intensive care units have the potential to influence the intestinal microbiome composition which may affect overall health status of the infant.
Collapse
Affiliation(s)
- Grace O Adeniyi-Ipadeola
- Graduate Program in Immunology & Microbiology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Kristi L Hoffman
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Heeju Yang
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Sara J Javornik Cregeen
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Geoffrey A Preidis
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Amy B Hair
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA.
| |
Collapse
|
12
|
Desorcy-Scherer K, Lamberti MFT, Weaver M, Lorca GL, Parker LA. Sociodemographic Factors and Intestinal Microbiome Development in Preterm, Very Low Birth Weight Infants. Am J Perinatol 2024; 41:e1866-e1877. [PMID: 37640050 DOI: 10.1055/s-0043-1769793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
OBJECTIVE Preterm very low birth weight (VLBW) infants are at risk for intestinal morbidities and dysbiotic development of the intestinal microbiome. Despite the influence of sociodemographic factors on premature infant health outcomes, whether they shape the intestinal microbiome early in life is not clear. The objective was to explore the associations between race, sex, and socioeconomic status and the intestinal microbiome of VLBW infants during the first 4 weeks of life. STUDY DESIGN This was a secondary analysis of data from an ongoing randomized trial of 79 infants ≤30 weeks' gestation and ≤1,500 g. Stool samples were collected at week 1 through week 4, frozen to -80°C and analyzed by 16S rRNA sequencing of the V4 region using Illumina MiSeq. Reads were analyzed to measure α and β diversity as well as relative abundance of bacteria in the intestinal microbiome. RESULTS Of the 79 infants, 63 had at least one sample available. Twenty-three (37%) of infants were African American, 30 (48%) were male, and 44 (71%) had Medicaid insurance. There were no statistically significant (<0.05) differences in α diversity or β diversity, and the differential abundance analysis suggests limited patterns of distinction in the intestinal microbiome between non-African American and African American infants, male and female infants, and infants with maternal private or Medicaid insurance. CONCLUSION Our results suggest race, sex, and socioeconomic status shape colonization of specific microorganisms to a limited extent. Future studies should confirm these findings and determine clinical relevance through further study of differentially abundant microorganisms and additional factors contributing to colonization patterns. KEY POINTS · Diversity of the gut microbiome was similar between infants of varying race, sex, and socioeconomic status.. · We observed sociodemographic-linked differences in colonization of individual taxa.. · Further study is required to confirm these results and the clinical relevance of these findings..
Collapse
Affiliation(s)
- Katelyn Desorcy-Scherer
- College of Nursing, University of Florida, Gainesville, Florida
- School of Nursing, University of Wisconsin, Madison, Wisconsin
| | - Monica F Torrez Lamberti
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, Florida
| | - Michael Weaver
- College of Nursing, University of Florida, Gainesville, Florida
| | - Graciela L Lorca
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, Florida
| | - Leslie A Parker
- College of Nursing, University of Florida, Gainesville, Florida
| |
Collapse
|
13
|
Wu D, Zhao P, Wang C, Huasai S, Chen H, Chen A. Differences in the intestinal microbiota and association of host metabolism with hair coat status in cattle. Front Microbiol 2024; 15:1296602. [PMID: 38711970 PMCID: PMC11071169 DOI: 10.3389/fmicb.2024.1296602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 03/11/2024] [Indexed: 05/08/2024] Open
Abstract
Introduction The hair coat status of cattle serves as an easily observed indicator of economic value in livestock production; however, the underlying mechanism remains largely unknown. Therefore, the objective of the current study was to determine differences in the intestinal microbiota and metabolome of cattle based on a division of with either slick and shining (SHC) or rough and dull (MHC) hair coat in Simmental cows. Methods Eight SHC and eight MHC late-pregnancy Simmental cows (with similar parities, body weights, and body conditions) were selected based on their hair coat status, and blood samples (plasma) from coccygeal venipuncture and fecal samples from the rectum were collected. The intestinal microbiota (in the fecal samples) was characterized by employing 16S rRNA gene sequencing targeting the V3-V4 hypervariable region on the Illumina MiSeq PE300 platform, and plasma samples were subjected to LC-MS/MS-based metabolomics with Progenesis QI 2.3. Plasma macromolecular metabolites were examined for differences in the metabolism of lipids, proteins, mineral elements, and hormones. Results Notable differences between the SHC and MHC groups related to host hair coat status were observed in the host metabolome and intestinal microbiota (P < 0.05). The host metabolome was enriched in histidine metabolism, cysteine and methionine metabolism, and purine metabolism in the SHC group, and the intestinal microbiota were also enriched in histidine metabolism (P < 0.05). In the MHC group, the symbiotic relationship transitioned from cooperation to competition in the MHC group, and an uncoupling effect was present in the microbe-metabolite association of intestine microbiota-host interactions. The hubs mediating the relationships between intestinal microbiota and plasma metabolites were the intestinal bacterial genus g__norank_f__Eubacterium_coprostanoligenes_group, plasma inosine, triiodothyronine, and phosphorus, which could be used to differentiate cows' hair coat status (P < 0.05). Conclusion Overall, the present study identified the relationships between the features of the intestinal microbiota and host hair coat status, thereby providing evidence and a new direction (intestine microbiota-host interplay) for future studies aimed at understanding the hair coat status of cattle.
Collapse
Affiliation(s)
- Donglin Wu
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Pengfei Zhao
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Chunjie Wang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Simujide Huasai
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Hao Chen
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Aorigele Chen
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
14
|
Parker LA, Koernere R, Fordham K, Bubshait H, Eugene A, Gefre A, Bendixen M. Mother's Own Milk Versus Donor Human Milk: What's the Difference? Crit Care Nurs Clin North Am 2024; 36:119-133. [PMID: 38296370 DOI: 10.1016/j.cnc.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Mother's own milk (MOM) is known to decrease complications in preterm infants and when unavailable, it is recommended that preterm very low-birth weight infants be fed donor human milk (DHM). Due to the pasteurization, processing, and lactation stage of donors, DHM does not contain the same nutritional, immunologic, and microbial components as MOM. This review summarizes the differences between MOM and DHM, the potential effects on health outcomes, and the clinical implications of these differences. Finally, implications for research and clinical practice are discussed.
Collapse
Affiliation(s)
- Leslie A Parker
- College of Nursing, University of Florida, Box 100187 College of Nursing, Gainesville, FL, USA.
| | - Rebecca Koernere
- College of Nursing, University of Florida, Box 100187 College of Nursing, Gainesville, FL, USA
| | - Keliy Fordham
- College of Nursing, University of Florida, Box 100187 College of Nursing, Gainesville, FL, USA
| | - Hussah Bubshait
- College of Nursing, University of Florida, Box 100187 College of Nursing, Gainesville, FL, USA
| | - Alissandre Eugene
- College of Nursing, University of Florida, Box 100187 College of Nursing, Gainesville, FL, USA
| | - Adrienne Gefre
- College of Nursing, University of Florida, Box 100187 College of Nursing, Gainesville, FL, USA
| | - Marion Bendixen
- College of Nursing, University of Florida, Box 100187 College of Nursing, Gainesville, FL, USA
| |
Collapse
|
15
|
Davis JA, Baumgartel K, Baust T, Conley YP, Morowitz MJ, Ren D, Demirci JR. Neonatal Diet Type and Associations With Adverse Feeding Outcomes in Neonates With Critical Congenital Heart Defects. J Perinat Neonatal Nurs 2024; 38:54-64. [PMID: 38236148 PMCID: PMC10807746 DOI: 10.1097/jpn.0000000000000790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 10/30/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Neonates with critical congenital heart defects (CCHD neonates) experience high rates of feeding intolerance, necrotizing enterocolitis (NEC), and malnutrition. The benefits of human milk and direct chest/breastfeeding are well known, but research is limited in CCHD neonates. Therefore, the purpose of this study is to examine the impact of neonatal diet and feeding modality on the incidence of feeding intolerance, NEC, and malnutrition among a cohort of CCHD neonates. METHODS A single-center retrospective study was conducted using electronic health record data of CCHD neonates admitted to a cardiac intensive care unit between April 2016 and April 2020. Regression models were fit to analyze associations between neonatal diet, feed modality, and adverse feeding outcomes. RESULTS Seventy-four CCHD neonates were included. Increased days of direct chest/breastfeeding were associated with fewer signs of gastrointestinal distress ( P = .047) and bloody stools ( P = .021). Enteral feeding days of "all human milk" were associated with higher growth trajectory ( P < .001). CONCLUSIONS Human milk and direct chest/breastfeeding may be protective against some adverse feeding outcomes for CCHD neonates. Larger, multicenter cohort studies are needed to continue investigating the effects of neonatal diet type and feeding modality on the development of adverse feeding outcomes in this unique population.
Collapse
Affiliation(s)
- Jessica A. Davis
- Division of General Academic Pediatrics, Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Davis); UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania (Drs Davis and Morowitz); School of Nursing, University of South Florida, Tampa (Dr Baumgartel); Departments of Critical Care Medicine (Ms Baust) and Surgery (Dr Morowitz), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and Departments of Health Promotion and Development (Dr Conley and Demirci) and Health and Community Systems (Dr Ren), School of Nursing, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kelley Baumgartel
- Division of General Academic Pediatrics, Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Davis); UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania (Drs Davis and Morowitz); School of Nursing, University of South Florida, Tampa (Dr Baumgartel); Departments of Critical Care Medicine (Ms Baust) and Surgery (Dr Morowitz), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and Departments of Health Promotion and Development (Dr Conley and Demirci) and Health and Community Systems (Dr Ren), School of Nursing, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Tracy Baust
- Division of General Academic Pediatrics, Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Davis); UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania (Drs Davis and Morowitz); School of Nursing, University of South Florida, Tampa (Dr Baumgartel); Departments of Critical Care Medicine (Ms Baust) and Surgery (Dr Morowitz), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and Departments of Health Promotion and Development (Dr Conley and Demirci) and Health and Community Systems (Dr Ren), School of Nursing, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yvette P. Conley
- Division of General Academic Pediatrics, Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Davis); UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania (Drs Davis and Morowitz); School of Nursing, University of South Florida, Tampa (Dr Baumgartel); Departments of Critical Care Medicine (Ms Baust) and Surgery (Dr Morowitz), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and Departments of Health Promotion and Development (Dr Conley and Demirci) and Health and Community Systems (Dr Ren), School of Nursing, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael J. Morowitz
- Division of General Academic Pediatrics, Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Davis); UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania (Drs Davis and Morowitz); School of Nursing, University of South Florida, Tampa (Dr Baumgartel); Departments of Critical Care Medicine (Ms Baust) and Surgery (Dr Morowitz), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and Departments of Health Promotion and Development (Dr Conley and Demirci) and Health and Community Systems (Dr Ren), School of Nursing, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Dianxu Ren
- Division of General Academic Pediatrics, Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Davis); UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania (Drs Davis and Morowitz); School of Nursing, University of South Florida, Tampa (Dr Baumgartel); Departments of Critical Care Medicine (Ms Baust) and Surgery (Dr Morowitz), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and Departments of Health Promotion and Development (Dr Conley and Demirci) and Health and Community Systems (Dr Ren), School of Nursing, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jill R. Demirci
- Division of General Academic Pediatrics, Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (Dr Davis); UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania (Drs Davis and Morowitz); School of Nursing, University of South Florida, Tampa (Dr Baumgartel); Departments of Critical Care Medicine (Ms Baust) and Surgery (Dr Morowitz), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and Departments of Health Promotion and Development (Dr Conley and Demirci) and Health and Community Systems (Dr Ren), School of Nursing, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
16
|
Bosco A, Piu C, Picciau ME, Pintus R, Fanos V, Dessì A. Metabolomics in NEC: An Updated Review. Metabolites 2023; 14:14. [PMID: 38248817 PMCID: PMC10821135 DOI: 10.3390/metabo14010014] [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: 11/11/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 01/23/2024] Open
Abstract
Necrotizing enterocolitis (NEC) represents the most common and lethal acute gastrointestinal emergency of newborns, mainly affecting those born prematurely. It can lead to severe long-term sequelae and the mortality rate is approximately 25%. Furthermore, the diagnosis is difficult, especially in the early stages, due to multifactorial pathogenesis and complex clinical pictures with mild and non-specific symptoms. In addition, the existing tests have poor diagnostic value. Thus, the scientific community has been focusing its attention on the identification of non-invasive biomarkers capable of prediction, early diagnosis and discriminating NEC from other intestinal diseases in order to intervene early and block the progression of the pathology. In this regard, the use of "omics" technologies, especially metabolomics and microbiomics, could be a fundamental synergistic strategy to study the pathophysiology of NEC. In addition, a deeper knowledge of the microbiota-host cross-talk can clarify the metabolic pathways potentially involved in the pathology, allowing for the identification of specific biomarkers. In this article, the authors analyze the state-of-the-art concerning the application of metabolomics and microbiota analysis to investigate this pathology and discuss the future possibility of the metabolomic fingerprint of patients for diagnostic purposes.
Collapse
Affiliation(s)
| | | | | | | | | | - Angelica Dessì
- Department of Surgical Sciences, University of Cagliari and Neonatal Intensive Care Unit, AOU Cagliari, 09124 Cagliari, Italy; (A.B.); (C.P.); (M.E.P.); (R.P.); (V.F.)
| |
Collapse
|
17
|
Bingham R, Pineda D, Gates A, Thompson AB, Stansfield BK. Displacement of human milk during fortification: An experimental study. JPEN J Parenter Enteral Nutr 2023; 47:1062-1066. [PMID: 37488685 DOI: 10.1002/jpen.2553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND This study quantified the displacement of human milk by commercial human milk fortifiers (HMFs) and infant formulas. METHODS Commercial liquid HMFs and powder infant formulas were added to pasteurized pooled donor human milk in triplicate, stirred, and weighed. The difference in weight between unfortified and fortified human milk at 22, 24, 26, 27, 28, and 30 kcal/ounce was calculated. RESULTS The displacement of human milk by liquid HMFs and powder infant formulas and powder HMF was highly associated with energy density. A human milk-derived HMF displaced significantly more human milk when compared with bovine milk-derived HMFs at equivalent energy densities. Similarly, powder infant formulas displaced less human milk when compared with a powder HMF, and the addition of hydrolyzed powder infant formulas resulted in less human milk displacement when compared with nonhydrolyzed powder infant formulas. CONCLUSIONS The displacement of human milk by commercial liquid HMFs and infant formulas must be considered when selecting a fortifying strategy.
Collapse
Affiliation(s)
- Rhyan Bingham
- Department of Pediatrics, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Daphne Pineda
- Department of Pediatrics, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Amy Gates
- Department of Pediatrics, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Amy B Thompson
- Department of Pediatrics, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Brian K Stansfield
- Department of Pediatrics, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| |
Collapse
|
18
|
Moreira DH, Gregory SB, Younge NE. Human milk fortification and use of infant formulas to support growth in the neonatal intensive care unit. Nutr Clin Pract 2023; 38 Suppl 2:S56-S65. [PMID: 37721458 PMCID: PMC10662944 DOI: 10.1002/ncp.11038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/03/2023] [Accepted: 06/10/2023] [Indexed: 09/19/2023] Open
Abstract
Newborn infants require adequate nutrition to achieve full potential growth and development. Early life nutrition and health impacts long-term outcomes through adulthood. Human milk is the optimal source of nutrition during the first 6 months of life. However, infants admitted to the neonatal intensive care unit (NICU) often have comorbidities that create more or different nutrition demands than healthy newborns. There are different strategies to meet the nutrition needs of sick newborns, including use of parenteral nutrition, human milk fortifiers (HMFs), and infant formulas. Multinutrient HMFs are frequently used to achieve the higher nutrition demands of preterm infants. They are available in various presentations, such as human milk- or cow milk-derived, liquid or powder, and acidified or nonacidified, each of which has different risks and benefits associated with its use. Infant formulas are available to meet a demand when mother's own milk or donor breast milk is not available or sufficient, and there are also specialty formulas for infants with certain diseases that present unique nutrition needs. This review is focused on the use of HMFs to support the unique nutrition requirements of preterm infants for healthy growth, as well as the indications for the use of formulas among infants in the NICU.
Collapse
Affiliation(s)
- Denise H Moreira
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Sarah B Gregory
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Noelle E Younge
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
19
|
Fu TT, Poindexter BB. Human Milk Fortification Strategies in the Neonatal Intensive Care Unit. Clin Perinatol 2023; 50:643-652. [PMID: 37536769 DOI: 10.1016/j.clp.2023.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
Multicomponent fortification is the standard of care to support short-term growth in preterm infants receiving human milk. There is no consensus regarding the optimal timing, method, or products used to fortify human milk. Both bovine milk-based and human milk-based human milk fortifiers are safe options, though increased fortification and enrichment may be needed to achieve adequate growth. Additional studies are needed to evaluate newer fortifier products and fortification strategies.
Collapse
Affiliation(s)
- Ting Ting Fu
- Division of Neonatology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Brenda B Poindexter
- Division of Neonatology, Department of Pediatrics, Children's Healthcare of Atlanta and Emory University School of Medicine, 2015 Uppergate Drive Northeast, Atlanta, GA 30322, USA
| |
Collapse
|
20
|
Liu K, Guo J, Yang J, Su Y. The Association of Different Proportions of Human Milk of the Total Enteral Intake on Health Outcomes in Preterm Infants: A Systematic Review. Breastfeed Med 2023; 18:666-677. [PMID: 37729036 DOI: 10.1089/bfm.2023.0128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Background: Human milk (HM) is a proven optimal food for preterm infants. However, there is uncertainty regarding the effects of different proportions of HM of the total enteral intake on health outcomes in preterm infants. Therefore, we conducted a systematic review of studies examining the effects of different proportions of HM of the total enteral intake on health outcomes in preterm infants. Methods: We conducted a literature search in the Web of Science, PubMed, and Scopus databases. The methodological quality of the included articles and the certainty of evidence were assessed according to the Newcastle-Ottawa Scale and the Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) tool, respectively. Results: Twelve studies were included in the review. Among the clinical findings, the effect of different proportions of HM of the total enteral feeding on health outcomes in preterm infants was divided into six primary outcomes: physical growth, length of stay, morbidity of any disease, all-cause mortality, feeding-related outcomes, and other health outcomes. The studies presented a high risk of bias for most of the domains. The certainty of the evidence was considered low or very low. Conclusions: The findings reiterated that greater proportions of HM positively affect the health outcomes of preterm infants. Overall, when the HM accounts for at least 20% of the total enteral intake, it has an effect on health outcomes in preterm infants. If the proportion of HM reaches 50%, the incidence and severity of necrotizing enterocolitis, as well as the time to reach enteral feeds, will be reduced. Increasing the proportion of HM in enteral feeding should be considered a priority in the feeding strategy for preterm infants in clinical practice.
Collapse
Affiliation(s)
- Keqin Liu
- School of Nursing, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinjin Guo
- School of Nursing, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jixin Yang
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanwei Su
- School of Nursing, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Yavanoglu Atay F, Bozkurt O, Sahin S, Bidev D, Sari FN, Uras N. A Comparison of Slow Infusion Intermittent Feeding versus Gravity Feeding in Preterm Infants: A Randomized Controlled Trial. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1389. [PMID: 37628387 PMCID: PMC10453490 DOI: 10.3390/children10081389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/01/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023]
Abstract
BACKGROUND The transition to full enteral feeding is important for ensuring adequate growth in preterm infants. AIMS The aim of this study was to investigate the effects of two different intermittent feeding methods on the transition to full enteral feeding in preterm infants. STUDY DESIGN A prospective, randomized controlled study was conducted in a neonatology and perinatology center. SUBJECTS Preterm infants with a gestational age between 24 + 0/7 and 31 + 6/7 were included in this study. They were divided into two groups: the SIF (slow infusion feeding) group and the IBF (intermittent bolus feeding) group. In the SIF group, feed volumes were administered over one hour using an infusion pump through an orogastric tube, with feeding occurring every three hours. The IBF group received enteral feeding using a gravity-based technique with a syringe through an orogastric tube, completed within 10 to 30 min. OUTCOME MEASURES The primary outcome was the achievement of full enteral feeding and the occurrence of feeding intolerance. RESULTS A total of 103 infants were enrolled in the study (50 in SIF and 53 in IBF). The time to achieve full enteral feeding did not differ significantly between the two groups (p = 0.20). The SIF group had significantly fewer occurrences in which gastric residual volume exceeded 50% (p = 0.01). Moreover, the SIF group had a significantly shorter duration of non-per-oral (NPO) status than the IBF group (p = 0.03). CONCLUSIONS It is our contention that the use of the SIF method as an alternative feeding method is appropriate for infants with feeding intolerance and those at high risk of feeding intolerance.
Collapse
Affiliation(s)
- Funda Yavanoglu Atay
- Department of Pediatrics, Division of Neonatology, Umraniye Training and Research Hospital, 34764 Istanbul, Türkiye
| | - Ozlem Bozkurt
- Department of Pediatrics, Division of Neonatology, Faculty of Medicine, Kocaeli University, 41380 Izmit, Türkiye
| | - Suzan Sahin
- Department of Pediatrics, Division of Neonatology, Buca Seyfi Demirsoy Training and Research Hospital, Izmir Democracy University, 35140 Izmir, Türkiye
| | - Duygu Bidev
- Neonatal Care Intensive Unit, Koru Sincan Hospital, 06934 Ankara, Türkiye
| | - Fatma Nur Sari
- Department of Pediatrics, Division of Neonatology, Ankara City Hospital, 06800 Ankara, Türkiye
| | - Nurdan Uras
- Department of Pediatrics, Division of Neonatology, Faculty of Medicine, Istinye University, 34517 Istanbul, Türkiye
| |
Collapse
|
22
|
Chen X, Shi Y. Determinants of microbial colonization in the premature gut. Mol Med 2023; 29:90. [PMID: 37407941 DOI: 10.1186/s10020-023-00689-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 06/20/2023] [Indexed: 07/07/2023] Open
Abstract
Abnormal microbial colonization in the gut at an early stage of life affects growth, development, and health, resulting in short- and long-term adverse effects. Microbial colonization patterns of preterm infants differ from those of full-term infants in that preterm babies and their mothers have more complicated prenatal and postnatal medical conditions. Maternal complications, antibiotic exposure, delivery mode, feeding type, and the use of probiotics may significantly shape the gut microbiota of preterm infants at an early stage of life; however, these influences subside with age. Although some factors and processes are difficult to intervene in or avoid, understanding the potential factors and determinants will help in developing timely strategies for a healthy gut microbiota in preterm infants. This review discusses potential determinants of gut microbial colonization in preterm infants and their underlying mechanisms.
Collapse
Affiliation(s)
- Xiaoyu Chen
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - Yongyan Shi
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110000, China.
| |
Collapse
|
23
|
Wu D, Zhang Z, Shao K, Wang X, Huang F, Qi J, Duan Y, Jia Y, Xu M. Effects of Sodium Butyrate Supplementation in Milk on the Growth Performance and Intestinal Microbiota of Preweaning Holstein Calves. Animals (Basel) 2023; 13:2069. [PMID: 37443869 DOI: 10.3390/ani13132069] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 06/10/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
The aim of the present study was to investigate the effects of sodium butyrate (SB) supplementation on the growth and intestinal microbiota of preweaning dairy calves. Eighty newborn Holstein calves (56 female and 24 male) were randomly allocated to four treatment groups with 20 calves each (14 female and 6 male). The suckling milk for the four treatments was supplemented with 0, 4.4, 8.8, or 17.6 g/d SB. During the 6-week experiment, dry matter intake was recorded daily, body weight was measured weekly, and rectal fecal samples were collected in the 2nd week. The V3-V4 hypervariable regions of the microbial 16S rRNA were amplified and then sequenced. SB supplementation elevated average daily gains (ADGs) in the first and second weeks. The optimal SB supplementation level for the whole preweaning period was 8.78 g/d, as revealed by analyzing the whole preweaning period ADG using second-order polynomial regression (quadratic) equations. The alpha diversity (Shannon diversity index), beta diversity, core phyla and genera, and function of the intestinal microbiota were affected by SB supplementation. In addition, the Shannon diversity index and core phyla and genera of the intestinal microbiota were correlated with calf growth-related indices. Overall, SB supplementation in suckling milk improved the growth performance and intestinal microbiota development of dairy calves in a quadratic manner, and regression analysis indicated an optimal supplementation level of 8.78 g/d.
Collapse
Affiliation(s)
- Donglin Wu
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Zhanhe Zhang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Kai Shao
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Xing Wang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Fudong Huang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Jingwei Qi
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Herbivorous Livestock Feed Engineering and Technology Research Center, Hohhot 010018, China
| | - Yizong Duan
- Shazhou Dairy Co., Ltd., Ulanqab 013750, China
| | - Yang Jia
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Ming Xu
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Herbivorous Livestock Feed Engineering and Technology Research Center, Hohhot 010018, China
| |
Collapse
|
24
|
Nicolas CT, Carter SR, Martin CA. Impact of maternal factors, environmental factors, and race on necrotizing enterocolitis. Semin Perinatol 2023; 47:151688. [PMID: 36572622 DOI: 10.1016/j.semperi.2022.151688] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Necrotizing enterocolitis (NEC) is a complex disease with a multifactorial etiology. As the leading cause of intestinal morbidity and mortality among premature infants, many resources are being dedicated to neonatal care and molecular targets in the newborn intestine. However, NEC is heavily influenced by maternal and perinatal factors as well. Given its nature, preventive approaches to NEC are more likely to improve outcomes than new treatment strategies. Therefore, this review focuses on maternal, environmental, and racial factors associated with the development of NEC, with an emphasis on those that may be modifiable to decrease the incidence of the disease.
Collapse
Affiliation(s)
- Clara T Nicolas
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Stewart R Carter
- Department of Surgery, Division of Pediatric Surgery, University of Louisville School of Medicine, Louisville, KY, United States
| | - Colin A Martin
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States; Department of General and Thoracic Surgery, Children's of Alabama, Birmingham, AL, United States.
| |
Collapse
|
25
|
Holzapfel LF, Hair AB, Preidis GA, Halder T, Yang H, Unger JP, Freedman S, Martin CR. Fecal Elastase in Preterm Infants to Predict Growth Outcomes. J Pediatr Gastroenterol Nutr 2023; 76:206-212. [PMID: 36705701 PMCID: PMC9886337 DOI: 10.1097/mpg.0000000000003672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVES Preterm infants are born functionally pancreatic insufficient with decreased pancreatic production of lipase and proteases. Developmental pancreatic insufficiency (PI) may contribute to reduced nutrient absorption and growth failure. We sought to determine longitudinal fecal elastase (ELA1) levels in a cohort of preterm infants and whether levels are associated with growth outcomes. METHODS Prospective observational study of 30 infants 24-34 weeks gestational age and birth weight ≤1250 g fed the exclusive human milk diet, consisting of human milk with human milk-based fortifier. ELA1 was quantified by ELISA during the first 2 weeks of life [Early; 7.5 ± 1.8 days of life (DOL)] and after attainment of full, fortified feedings (Late; 63.6 ± 24.1 DOL). RESULTS Early ELA1 levels were 192.2 ± 96.4 µg/g, and Late ELA1 levels were 268.0 ± 80.3 µg/g, 39.4% higher (P = 0.01). Infants with early PI (ELA1 < 200 µg/g) were more likely male and of lower gestational age, weight, length, and head circumference at birth. These variables, but not PI status, independently predicted somatic growth. CONCLUSIONS Fecal ELA1 in preterm infants fed exclusive human milk diet increases with postnatal age. Although pancreatic function in preterm infants may serve as a biological contributor to early postnatal growth failure, additional studies using fecal ELA1 as a predictive biomarker for growth failure are needed in larger cohorts.
Collapse
Affiliation(s)
- Lindsay F Holzapfel
- From the Department of Pediatrics, Division of Neonatology, University of Texas at Houston Health Science Center, Houston, TX
- the Department of Pediatrics, Section of Neonatology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX
| | - Amy B Hair
- the Department of Pediatrics, Section of Neonatology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX
| | - Geoffrey A Preidis
- the Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Tripti Halder
- the Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Heeju Yang
- the Department of Pediatrics, Section of Neonatology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX
| | - Jana P Unger
- the Department of Pediatrics, Section of Neonatology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX
- the Clinical Nutrition Services, Texas Children's Hospital, Houston, TX
| | - Steven Freedman
- the Department of Gastroenterology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
- Division of Translational Research, Beth Israel Deaconess Medical Center, Boston, MA
| | - Camilia R Martin
- the Division of Neonatology, Weill Cornell Medicine, New York, NY
| |
Collapse
|
26
|
Neves LL, Hair AB, Preidis GA. A systematic review of associations between gut microbiota composition and growth failure in preterm neonates. Gut Microbes 2023; 15:2190301. [PMID: 36927287 PMCID: PMC10026866 DOI: 10.1080/19490976.2023.2190301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
Growth failure is among the most prevalent and devastating consequences of prematurity. Up to half of all extremely preterm neonates struggle to grow despite modern nutrition practices. Although elegant preclinical models suggest causal roles for the gut microbiome, these insights have not yet translated into biomarkers that identify at-risk neonates or therapies that prevent or treat growth failure. This systematic review aims to identify features of the neonatal gut microbiota that are positively or negatively associated with early postnatal growth. We identified 860 articles, of which 14 were eligible for inclusion. No two studies used the same definitions of growth, ages at stool collection, and statistical methods linking microbiota to metadata. In all, 58 different taxa were associated with growth, with little consensus among studies. Two or more studies reported positive associations with Enterobacteriaceae, Bacteroides, Bifidobacterium, Enterococcus, and Veillonella, and negative associations with Citrobacter, Klebsiella, and Staphylococcus. Streptococcus was positively associated with growth in five studies and negatively associated with growth in three studies. To gain insight into how the various definitions of growth could impact results, we performed an exploratory secondary analysis of 245 longitudinally sampled preterm infant stools, linking microbiota composition to multiple clinically relevant definitions of neonatal growth. Within this cohort, every definition of growth was associated with a different combination of microbiota features. Together, these results suggest that the lack of consensus in defining neonatal growth may limit our capacity to detect consistent, meaningful clinical associations that could be leveraged into improved care for preterm neonates.
Collapse
Affiliation(s)
- Larissa L. Neves
- Division of Gastroenterology, Hepatology, & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Amy B. Hair
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Geoffrey A. Preidis
- Division of Gastroenterology, Hepatology, & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| |
Collapse
|
27
|
Lynch LE, Hair AB, Soni KG, Yang H, Gollins LA, Narvaez-Rivas M, Setchell KDR, Preidis GA. Cholestasis impairs gut microbiota development and bile salt hydrolase activity in preterm neonates. Gut Microbes 2023; 15:2183690. [PMID: 36843227 PMCID: PMC9980517 DOI: 10.1080/19490976.2023.2183690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/28/2023] Open
Abstract
Cholestasis refers to impaired bile flow from the liver to the intestine. In neonates, cholestasis causes poor growth and may progress to liver failure and death. Normal bile flow requires an intact liver-gut-microbiome axis, whereby liver-derived primary bile acids are transformed into secondary bile acids. Microbial bile salt hydrolase (BSH) enzymes are responsible for the first step, deconjugating glycine- and taurine-conjugated primary bile acids. Cholestatic neonates often are treated with the potent choleretic bile acid ursodeoxycholic acid (UDCA), although interactions between UDCA, gut microbes, and other bile acids are poorly understood. To gain insight into how the liver-gut-microbiome axis develops in extreme prematurity and how cholestasis alters this maturation, we conducted a nested case-control study collecting 124 stool samples longitudinally from 24 preterm infants born at mean 27.2 ± 1.8 weeks gestation and 946 ± 249.6 g, half of whom developed physiologic cholestasis. Samples were analyzed by whole metagenomic sequencing, in vitro BSH enzyme activity assays optimized for low biomass fecal samples, and quantitative mass spectrometry to measure the bile acid metabolome. In extremely preterm neonates, acquisition of the secondary bile acid biosynthesis pathway and BSH genes carried by Clostridium perfringens are the most prominent features of early microbiome development. Cholestasis interrupts this developmental pattern. BSH gene abundance and enzyme activity are profoundly reduced in cholestatic neonates, resulting in decreased quantities of unconjugated bile acids. UDCA restores total fecal bile acid levels in cholestatic neonates, but this is due to a 522-fold increase in fecal UDCA. A majority of bile acids in early development are atypical positional and stereo-isomers of bile acids. We report novel associations linking isomeric bile acids and BSH activity to neonatal growth trajectories. These data highlight deconjugation of bile acids as a key microbial function that is acquired in early neonatal development and impaired by cholestasis.
Collapse
Affiliation(s)
- Lauren E. Lynch
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Amy B. Hair
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA,CONTACT Amy B. Hair Division of Neonatology, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, 6621 Fannin Street, Suite A5590, Houston, TX77030, USA
| | - Krishnakant G. Soni
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Heeju Yang
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Laura A. Gollins
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Monica Narvaez-Rivas
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Kenneth D. R. Setchell
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Geoffrey A. Preidis
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA,Geoffrey A. Preidis Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, 1102 Bates Avenue, Feigin Tower Suite 860, Houston, TX77030, USA
| |
Collapse
|
28
|
Differences in Neonatal Outcomes Among Premature Infants Exposed to Mother's Own Milk Versus Donor Human Milk. Adv Neonatal Care 2022; 22:539-549. [PMID: 35939792 DOI: 10.1097/anc.0000000000001002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Growing evidence supports the superior benefits of exposure to mother's own milk (MOM) in reducing prematurity-related comorbidities. Neonatal exposure to donor human Milk (DHM) is a suitable alternative when MOM is insufficient or unavailable. However, the same protective composition and bioactivity in MOM are not present in DHM. Additional evidence is needed to justify and inform evidence-based practices increasing MOM provision while optimizing adequate use of DHM for premature infants. PURPOSE A systematic review of the literature was conducted to determine differences in neonatal outcomes among premature infants exposed to predominately MOM versus DHM. METHODS/SEARCH STRATEGY Databases including PubMed, CINAHL and Cochrane were searched (2020-2021) using the PRISMA (Preferred Reporting Items for Systematic reviews and Meta-Analysis) guidelines. Evidence was classified using the John Hopkins evidence-based practice levels and quality of evidence. RESULTS Eleven studies met inclusion criteria. Studied neonatal outcomes included ( a ) growth parameters (n = 8), ( b ) neonatal morbidities (n = 6), and ( c ) gut microbiome (n = 4). Overall, evidence suggests DHM exposure is beneficial but not equivalent to MOM feeding. Compared with DHM, greater doses of MOM are ideal to enhance protection primarily related to infant growth, as well as gut microbiome diversity and richness. IMPLICATIONS FOR PRACTICE Standardized and evidence-based practices are needed to clearly delineate optimal use of DHM without undermining maternal and neonatal staff efforts to support and promote provision of MOM. IMPLICATIONS FOR RESEARCH Additional evidence from high-quality studies should further examine differences in neonatal outcomes among infants exposed to predominately MOM or DHM in settings using standardized and evidence-based feeding practices.
Collapse
|
29
|
Sarkar A, Prescott SM, Dutra S, Yoo JY, Gordon J, Shaffer E, McSkimming D, Groer ME. Relationships of the very low birth weight infant microbiome with neurodevelopment at 2 and 4 years of age. Dev Psychobiol 2022; 64:e22317. [PMID: 36282736 PMCID: PMC9608354 DOI: 10.1002/dev.22317] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/17/2022] [Accepted: 07/18/2022] [Indexed: 01/27/2023]
Abstract
Very low birth weight (VLBW) infants (<1500 g) are at risk for poor neurodevelopmental outcomes depending on gestational age (GA), birth weight (BW), and morbidity in early life. The contribution of the gut microbiome is not well understood. Stool samples were collected weekly in the neonatal intensive care unit (NICU) from 24 VLBW infants for 6 weeks after admission and then again at 2 and 4 years of age. The Battelle Development Inventory-2 Screening Test (BDI-2 ST) was administered at 2- and 4-year time points. VLBW infants had dysbiotic microbiota in the NICU that progressed for most to an adult-type microbiota by 4 years of age. The BDI-2 ST results at age of 2 years triggered referral for further testing in 14 toddlers (70%), and by 4 years of age only seven of these 14 continued to require referral. Both NICU infant stool diversity and particular microbial amplicon sequence variants were associated with BDI-2 ST subscales, particularly for cognition, adaptive, and communication subscales, when controlled for GA, BW, and antibiotic exposure. Network analysis of the NICU infant stool microbial ecology showed differences in children needing neurodevelopmental referral. The results of this preliminary study indicate that the neonatal gut microbiome plays a role in early cognitive and behavioral neurodevelopment.
Collapse
Affiliation(s)
- Anujit Sarkar
- University of South Florida College of Nursing, Tampa, Florida, USA
- University of South Florida College of Public Health, Tampa, Florida, USA
| | - Stephanie M Prescott
- University of South Florida College of Nursing, Tampa, Florida, USA
- Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Samia Dutra
- University of South Florida College of Nursing, Tampa, Florida, USA
| | - Ji Youn Yoo
- University of Tennessee Knoxville College of Nursing, Knoxville, Tennessee, USA
| | - Jessica Gordon
- University of South Florida College of Nursing, Tampa, Florida, USA
| | - Emily Shaffer
- University of South Florida College of Public Health, Tampa, Florida, USA
| | - Daniel McSkimming
- Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Maureen E Groer
- University of South Florida College of Nursing, Tampa, Florida, USA
- Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
30
|
Impact of Donor Human Milk in an Urban NICU Population. CHILDREN (BASEL, SWITZERLAND) 2022; 9:children9111639. [PMID: 36360367 PMCID: PMC9688816 DOI: 10.3390/children9111639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
Abstract
The American Academy of Pediatrics recommends the use of donor human milk in infants when mother’s own milk is not available. Our objective was to analyze whether the use of donor human milk in preterm, very-low-birth-weight (VLBW, <1500 g) infants affected the rates of necrotizing enterocolitis, duration of parenteral nutrition (PN), growth, culture-positive sepsis, length of hospital stay, and mortality in an urban NICU population with low exclusive breast-feeding rates. A retrospective cohort study was conducted comparing two 2-year epochs of VLBW neonates before and after the introduction of donor breast milk in our neonatal intensive care unit (NICU). With the introduction of donor human milk, there was a significant reduction in the rate of necrotizing enterocolitis (NEC) (5% vs. 13%; p = 0.04) and less severe NEC as defined by Stage III based on the Modified Bell Staging Criteria (10% to 3%; p = 0.04). In the donor milk era, there was earlier initiation of enteral feeding (2.69 days vs. 3.84; p = 0.006) and a more rapid return to birthweight (9.5 days. 10.9 days; p = 0.006). In this study, a change in practice to the use of donor breast milk in a population with low rates of human milk provision was associated with earlier initiation of enteral feeding, faster return to birth weight, and a reduced incidence of NEC.
Collapse
|
31
|
Kumbhare SV, Jones WD, Fast S, Bonner C, Jong G', Van Domselaar G, Graham M, Narvey M, Azad MB. Source of human milk (mother or donor) is more important than fortifier type (human or bovine) in shaping the preterm infant microbiome. Cell Rep Med 2022; 3:100712. [PMID: 36029771 PMCID: PMC9512671 DOI: 10.1016/j.xcrm.2022.100712] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/13/2022] [Accepted: 07/14/2022] [Indexed: 12/20/2022]
Abstract
Milk fortifiers help meet the nutritional needs of preterm infants receiving their mother's own milk (MOM) or donor human milk. We conducted a randomized clinical trial (NCT03214822) in 30 very low birth weight premature neonates comparing bovine-derived human milk fortifier (BHMF) versus human-derived fortifier (H2MF). We found that fortifier type does not affect the overall microbiome, although H2MF infants were less often colonized by an unclassified member of Clostridiales Family XI. Secondary analyses show that MOM intake is strongly associated with weight gain and microbiota composition, including Bifidobacterium, Veillonella, and Propionibacterium enrichment. Finally, we show that while oxidative stress (urinary F2-isoprostanes) is not affected by fortifier type or MOM intake, fecal calprotectin is higher in H2MF infants and lower in those consuming more MOM. Overall, the source of human milk (mother versus donor) appears more important than the type of milk fortifier (human versus bovine) in shaping preterm infant gut microbiota.
Collapse
Affiliation(s)
- Shreyas V Kumbhare
- Manitoba Interdisciplinary Lactation Centre (MILC), Winnipeg, MB, Canada; Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB, Canada; Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - William-Diehl Jones
- Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB, Canada; Faculty of Health Disciplines, Athabasca University, Athabasca, AB, Canada
| | - Sharla Fast
- Department of Nutrition and Food Services, Health Sciences Centre, Winnipeg, MB, Canada
| | - Christine Bonner
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Geert 't Jong
- Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB, Canada; Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Gary Van Domselaar
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada; Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Morag Graham
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada; Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Michael Narvey
- Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB, Canada; Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Meghan B Azad
- Manitoba Interdisciplinary Lactation Centre (MILC), Winnipeg, MB, Canada; Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB, Canada; Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
32
|
Albraik RK, Shatla E, Abdulla YM, Ahmed EH. Neonatal Feeding Intolerance and Its Characteristics: A Descriptive Study. Cureus 2022; 14:e29291. [PMID: 36277537 PMCID: PMC9578381 DOI: 10.7759/cureus.29291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2022] [Indexed: 11/05/2022] Open
|
33
|
Asbury MR, Shama S, Sa JY, Bando N, Butcher J, Comelli EM, Copeland JK, Forte V, Kiss A, Sherman PM, Stintzi A, Taibi A, Tomlinson C, Unger S, Wang PW, O'Connor DL. Human milk nutrient fortifiers alter the developing gastrointestinal microbiota of very-low-birth-weight infants. Cell Host Microbe 2022; 30:1328-1339.e5. [PMID: 35987195 DOI: 10.1016/j.chom.2022.07.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/14/2022] [Accepted: 07/15/2022] [Indexed: 11/30/2022]
Abstract
Nutrient fortifiers are added to human milk to support the development of very-low-birth-weight infants. Currently, bovine-milk-based fortifiers (BMBFs) are predominantly administered, with increasing interest in adopting human-milk-based fortifiers (HMBFs). Although beneficial for growth, their effects on the gastrointestinal microbiota are unclear. This triple-blind, randomized clinical trial (NCT02137473) tested how nutrient-enriching human milk with HMBF versus BMBF affects the gastrointestinal microbiota of infants born < 1,250 g during hospitalization. HMBF-fed infants (n = 63, n = 269 stools) showed lower microbial diversity, altered microbial community structure, and changes in predicted microbial functions compared with BMBF-fed infants (n = 56, n = 239 stools). HMBF-fed infants had higher relative and normalized abundances of unclassified Enterobacteriaceae and lower abundances of Clostridium sensu stricto. Post hoc analyses identified dose-dependent relationships between individual feed components (volumes of mother's milk, donor milk, and fortifiers) and the microbiota. These results highlight how nutrient fortifiers impact the microbiota of very-low-birth-weight infants during a critical developmental window.
Collapse
Affiliation(s)
- Michelle R Asbury
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Sara Shama
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Jong Yup Sa
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Nicole Bando
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - James Butcher
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1N 6N5, Canada; Ottawa Institute of Systems Biology, University of Ottawa, Ottawa ON, K1H 8M5, Canada
| | - Elena M Comelli
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Joannah and Brian Lawson Centre for Child Nutrition, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Julia K Copeland
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, ON M5S 3B2, Canada
| | - Victoria Forte
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Alex Kiss
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON M5T 3M6, Canada; Evaluative and Clinical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Philip M Sherman
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Cell Biology Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1X3, Canada
| | - Alain Stintzi
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1N 6N5, Canada; Ottawa Institute of Systems Biology, University of Ottawa, Ottawa ON, K1H 8M5, Canada
| | - Amel Taibi
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Christopher Tomlinson
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Joannah and Brian Lawson Centre for Child Nutrition, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Pediatrics, University of Toronto, Toronto, ON M5G 1X8, Canada; Division of Neonatology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Sharon Unger
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Pediatrics, University of Toronto, Toronto, ON M5G 1X8, Canada; Division of Neonatology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Department of Pediatrics, Sinai Health, Toronto, ON M5G 1X5, Canada; Rogers Hixon Ontario Human Milk Bank, Toronto, ON M5G 1X5, Canada
| | - Pauline W Wang
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, ON M5S 3B2, Canada
| | - Deborah L O'Connor
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Translational Medicine Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Joannah and Brian Lawson Centre for Child Nutrition, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Pediatrics, Sinai Health, Toronto, ON M5G 1X5, Canada; Rogers Hixon Ontario Human Milk Bank, Toronto, ON M5G 1X5, Canada.
| | | |
Collapse
|
34
|
Meier PP. More evidence: Mothers’ own milk is personalized medicine for very low birthweight infants. Cell Rep Med 2022; 3:100710. [PMID: 35977466 PMCID: PMC9418736 DOI: 10.1016/j.xcrm.2022.100710] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
“Human milk sources and fortification: Impact on fecal microbiota and calprotectin in premature infants” highlights improved outcomes in predominantly mothers’ own milk-fed versus predominantly pasteurized donor human milk-fed VLBW infants, regardless of fortifier type. Research and practice implications are reviewed.
Collapse
|
35
|
Waltz P, Azzuqa A. Nutritional considerations in the neonate. Semin Pediatr Surg 2022; 31:151197. [PMID: 36038219 DOI: 10.1016/j.sempedsurg.2022.151197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Paul Waltz
- Assistant Professor of Surgery, UPMC Childrens Hospital of Pittsburgh, USA.
| | - Abeer Azzuqa
- Associate Professor of Pediatrics, UPMC Childrens Hospital of Pittsburgh, USA
| |
Collapse
|
36
|
Balasundaram M, Land R, Miller S, Profit J, Porter M, Arnold C, Sivakumar D. Increasing early exposure to mother's own milk in premature newborns. J Perinatol 2022; 42:1126-1134. [PMID: 35396577 DOI: 10.1038/s41372-022-01376-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/02/2022] [Accepted: 03/17/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Increase the proportion of ≤33 weeks newborns exposed to mother's own milk (MOM) oral care by 12 h of age by 20% over 2 years to support a healthier microbiome. STUDY DESIGN We implemented interventions to support early expression of colostrum and reliable delivery of resultant MOM to premature newborns. Statistical process control charts were used to track progress and provide feedback to staff. Proportions of newborns exposed to MOM by 12 h were compared relative to baseline. RESULTS There were 46, 66, and 46 newborns in the baseline, implementation, and sustainability periods, respectively. The primary outcome improved from 48% to 61% in the implementation period (relative change 1.27, 95% CI 0.89, 1.81, p = 0.2), to 69% in sustainability period (relative to baseline 1.45, 95% CI 1.02, 2.08, p = 0.03). CONCLUSION An interdisciplinary team-based, multicycle, quality improvement intervention resulted in increased rates of early exposure to MOM.
Collapse
Affiliation(s)
- Malathi Balasundaram
- Division of Neonatal & Developmental Medicine, Stanford University School of Medicine, Palo Alto, CA, USA. .,Neonatal Intensive Care Unit, El Camino Health, Mountain View, CA, USA.
| | - Rachel Land
- Division of Neonatal & Developmental Medicine, Stanford University School of Medicine, Palo Alto, CA, USA.,Neonatal Intensive Care Unit, El Camino Health, Mountain View, CA, USA
| | - Stephanie Miller
- Division of Neonatal & Developmental Medicine, Stanford University School of Medicine, Palo Alto, CA, USA.,Neonatal Intensive Care Unit, El Camino Health, Mountain View, CA, USA
| | - Jochen Profit
- Division of Neonatal & Developmental Medicine, Stanford University School of Medicine, Palo Alto, CA, USA.,California Perinatal Quality Care Collaborative, Palo Alto, CA, USA
| | - Melinda Porter
- Neonatal Intensive Care Unit, El Camino Health, Mountain View, CA, USA
| | - Cody Arnold
- Division of Neonatal & Developmental Medicine, Stanford University School of Medicine, Palo Alto, CA, USA.,Neonatal Intensive Care Unit, El Camino Health, Mountain View, CA, USA
| | - Dharshi Sivakumar
- Division of Neonatal & Developmental Medicine, Stanford University School of Medicine, Palo Alto, CA, USA.,Neonatal Intensive Care Unit, El Camino Health, Mountain View, CA, USA
| |
Collapse
|
37
|
Wickland J, Wade C, Micetic B, Meredith K, Martin G. A Retrospective Analysis of the Effects of an Exclusively Human Milk Protein Diet on Neonatal Feeding Tolerance. Am J Perinatol 2022; 39:995-1000. [PMID: 33249551 DOI: 10.1055/s-0040-1721374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVE This study was aimed to evaluate the effect of human milk protein fortifier (HMPF) versus bovine milk protein fortifier (BMPF) on feeding tolerance defined as the time to reach full feeds and necrotizing enterocolitis (NEC) in premature very low birth weight (VLBW) infants. STUDY DESIGN A retrospective review using the BabySteps Database included 493 infants born ≤33 weeks of gestational age and ≤1,250 g (g) birth weight. A total of 218 infants fed a human milk diet (HMD) with BMPF were compared with 275 infants fed an HMD with HMPF. RESULTS Full feeds were reached significantly sooner in the HMPF group (median: 14 vs. 16 days, p = 0.04). Weight at full feeds was significantly lower in the HMPF group (1,060 vs. 1110 g, p = 0.03). CONCLUSION Using HMPF to provide an exclusively HMD allowed VLBW infants to achieve full feeds sooner, but did not affect rate of NEC compared with using a BMPF with an HMD. KEY POINTS · Human milk fortifiers with human milk are better than bovine human milk protein fortifiers.. · Full feeds are reached sooner with a human milk protein fortifier.. · The incidence of NEC did not change with the use of Prolacta..
Collapse
Affiliation(s)
- Jessica Wickland
- Division of Neonatal Perinatal Medicine, Department of Pediatrics, UT Southwestern Medical Center, Dallas, Texas.,Division of Neonatology, Phoenix Children's Medical Group, Phoenix, Arizona
| | - Christine Wade
- Mednax Services Inc./Phoenix Perinatal Associates, Phoenix, Arizona
| | - Becky Micetic
- Mednax Services Inc./Phoenix Perinatal Associates, Phoenix, Arizona
| | - Keith Meredith
- Mednax Services Inc./Phoenix Perinatal Associates, Phoenix, Arizona
| | - Gregory Martin
- Division of Neonatology, Phoenix Children's Medical Group, Phoenix, Arizona.,Mednax Services Inc./Phoenix Perinatal Associates, Phoenix, Arizona
| |
Collapse
|
38
|
Shen W, Qiu W, Lin Q, Zeng C, Liu Y, Huang W, Zhou H. The Gut Microbiome of Preterm Infants Treated With Aminophylline Is Closely Related to the Occurrence of Feeding Intolerance and the Weight Gain. Front Nutr 2022; 9:905839. [PMID: 35719163 PMCID: PMC9198222 DOI: 10.3389/fnut.2022.905839] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 04/29/2022] [Indexed: 11/21/2022] Open
Abstract
Background Aminophylline is widely used in the treatment of preterm infants, but it can cause feeding intolerance events, in which gut microbial dysbiosis may have a role. This study aims to investigate the relationship between the gut microbiome of preterm infants treated with aminophylline and the occurrence of feeding intolerance and weight gain rate. Methods This study included a cohort of 118 preterm infants. Survival analysis and multivariate Cox regression were used to evaluate the relationship between aminophylline treatment and the occurrence of feeding intolerance. 16S rRNA V4 region gene sequencing was used to characterize the microbiome of fecal samples from the cohort. Linear discriminant analysis effect size was used to analyze the differential abundance of bacteria related to aminophylline treatment. Wilcoxon test, Kruskal–Wallis test, Spearman correlation coefficients and generalized linear mixed models were used to analyze the correlation between the differential bacteria and feeding intolerance events as well as the weight gain. Results The results showed that the use of aminophylline could significantly increase the occurrence of feeding intolerance. The relative abundances of Streptococcus and Rothia in the gut microbiome of preterm infants were positively correlated with both the occurrence of feeding intolerance and the use of aminophylline, while the relative abundance of Staphylococcus was negatively correlated. In particular, preterm infants with a lower relative abundance of Rothia were more likely to develop feeding intolerance associated with aminophylline, and this difference existed before the onset of feeding intolerance. Moreover, it took longer for individuals with a lower relative abundance of Streptococcus to reach 2 kg weight. The contribution of Streptococcus to weight gain was greater than that of Bifidobacterium or Lactobacillus. Conclusion The gut microbiome in preterm infants treated with aminophylline was characterized by a decrease in Streptococcus and Rothia and an increase in Staphylococcus. These microbes, especially Rothia, were positively correlated with the occurrence of feeding intolerance. Streptococcus but not Bifidobacter likely participated in the weight gain of preterm infants in early life.
Collapse
Affiliation(s)
- Wei Shen
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wen Qiu
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qi Lin
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chao Zeng
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuting Liu
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weimin Huang
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
39
|
Engevik MA, Stripe LK, Baatz JE, Wagner CL, Chetta KE. Identifying single-strain growth patterns of human gut microbes in response to preterm human milk and formula. Food Funct 2022; 13:5571-5589. [PMID: 35481924 DOI: 10.1039/d2fo00447j] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The intestinal microbiota of the preterm neonate has become a major research focus, with evidence emerging that the microbiota influences both short and long-term health outcomes, in the neonatal intensive care unit and beyond. Similar to the term microbiome, the preterm gut microbiome is highly influenced by diet, specifically formula and human milk use. This study aims to analyze next-generation products including preterm formula, human milk-oligosaccharide term formula, and preterm breastmilk. We used a culture-based model to differentially compare the growth patterns of individual bacterial strains found in the human intestine. This model probed 24 strains of commensal bacteria and 8 pathobiont species which have previously been found to cause sepsis in preterm neonates. Remarkable differences between strain growth and culture pH were noted after comparing models of formulas and between human milk and formula. Both formula and human milk supported the growth of commensal bacteria; however, the formula products, but not human milk, supported the growth of several specific pathogenic strains. Computational analysis revealed potential connections between long-chain fatty acid and iron uptake from formula in pathobiont organisms. These findings indicate that there is a unique profile of growth in response to human milk and formula and shed light into how the infant gut microbiota could be influenced.
Collapse
Affiliation(s)
- Melinda A Engevik
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina, USA.,Department of Microbiology & Immunology, Medical University of South Carolina, USA
| | - Leah K Stripe
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina, USA
| | - John E Baatz
- Department of Pediatrics, C.P. Darby Children's Research Institute, Medical University of South Carolina, USA. .,Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Medical University of South Carolina, Shawn Jenkins Children's Hospital, 10 McClennan Banks Drive, MSC 915, Charleston, SC 29425, USA
| | - Carol L Wagner
- Department of Pediatrics, C.P. Darby Children's Research Institute, Medical University of South Carolina, USA. .,Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Medical University of South Carolina, Shawn Jenkins Children's Hospital, 10 McClennan Banks Drive, MSC 915, Charleston, SC 29425, USA
| | - Katherine E Chetta
- Department of Pediatrics, C.P. Darby Children's Research Institute, Medical University of South Carolina, USA. .,Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Medical University of South Carolina, Shawn Jenkins Children's Hospital, 10 McClennan Banks Drive, MSC 915, Charleston, SC 29425, USA
| |
Collapse
|
40
|
Yu X, Xing Y, Liu H, Chang Y, You Y, Dou Y, Liu B, Wang Q, Ma D, Chen L, Tong X. Effects of a Formula with scGOS/lcFOS (9:1) and Glycomacropeptide (GMP) Supplementation on the Gut Microbiota of Very Preterm Infants. Nutrients 2022; 14:nu14091901. [PMID: 35565868 PMCID: PMC9102166 DOI: 10.3390/nu14091901] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/20/2022] [Accepted: 04/27/2022] [Indexed: 01/27/2023] Open
Abstract
Microbial colonization of very preterm (VPT) infants is detrimentally affected by the complex interplay of physiological, dietary, medical, and environmental factors. The aim of this study was to evaluate the effects of an infant formula containing the specific prebiotic mixture of scGOS/lcFOS (9:1) and glycomacropeptide (GMP) on the composition and function of VPT infants’ gut microbiota. Metagenomic analysis was performed on the gut microbiota of VPT infants sampled at four time points: 24 h before the trial and 7, 14, and 28 days after the trial. Functional profiling was aggregated into gut and brain modules (GBMs) and gut metabolic modules (GMMs) based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Enterococcus faecium, Escherichia coli, Klebsiella aerogenes, and Klebsiella pneumoniae were dominant species in both the test group and the control group. After the 4-week intervention, the abundance of Bifidobacterium in the test group was significantly increased. We found two GBMs (quinolinic acid synthesis and kynurenine degradation) and four GMMs (glutamine degradation, glyoxylate bypass, dissimilatory nitrate reduction, and preparatory phase of glycolysis) were significantly enriched in the test group, respectively. The results of this study suggested that formula enriched with scGOS/lcFOS (9:1) and GPM is beneficial to the intestinal microecology of VPT infants.
Collapse
Affiliation(s)
- Xue Yu
- School of Public Health, Peking University Health Science Center, Beijing 100191, China; (X.Y.); (Y.D.)
| | - Yan Xing
- Department of Pediatrics, Peking University Third Hospital, Beijing 100191, China; (Y.X.); (H.L.); (Y.C.); (Y.Y.)
| | - Hui Liu
- Department of Pediatrics, Peking University Third Hospital, Beijing 100191, China; (Y.X.); (H.L.); (Y.C.); (Y.Y.)
| | - Yanmei Chang
- Department of Pediatrics, Peking University Third Hospital, Beijing 100191, China; (Y.X.); (H.L.); (Y.C.); (Y.Y.)
| | - Yanxia You
- Department of Pediatrics, Peking University Third Hospital, Beijing 100191, China; (Y.X.); (H.L.); (Y.C.); (Y.Y.)
| | - Yuqi Dou
- School of Public Health, Peking University Health Science Center, Beijing 100191, China; (X.Y.); (Y.D.)
| | - Bin Liu
- National Engineering Center of Dairy for Maternal and Child Health, Beijing Sanyuan Foods Co., Ltd., Beijing 100163, China;
| | - Qi Wang
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730030, China;
| | - Defu Ma
- School of Public Health, Peking University Health Science Center, Beijing 100191, China; (X.Y.); (Y.D.)
- Correspondence: (D.M.); (L.C.); (X.T.)
| | - Lijun Chen
- National Engineering Center of Dairy for Maternal and Child Health, Beijing Sanyuan Foods Co., Ltd., Beijing 100163, China;
- Correspondence: (D.M.); (L.C.); (X.T.)
| | - Xiaomei Tong
- Department of Pediatrics, Peking University Third Hospital, Beijing 100191, China; (Y.X.); (H.L.); (Y.C.); (Y.Y.)
- Correspondence: (D.M.); (L.C.); (X.T.)
| |
Collapse
|
41
|
Elgersma KM, McKechnie AC, Schorr EN, Shah KM, Trebilcock AL, Ramel SE, Ambrose MB, Swanson NM, Sommerness SA, Spatz DL. The Impact of Human Milk on Outcomes for Infants with Congenital Heart Disease: A Systematic Review. Breastfeed Med 2022; 17:393-411. [PMID: 35167760 DOI: 10.1089/bfm.2021.0334] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background: Infants with congenital heart disease (CHD) are at risk for feeding-related morbidity and mortality, with growth failure and oral feeding problems associated with poor outcomes. The benefits of human milk (HM) for preterm infants have been well documented, but evidence on HM for infants with CHD has recently begun to emerge. Objectives: Our primary aim was to examine the impact of HM feeding on outcomes for infants with CHD. Methods: Following PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-analyses) guidelines, a search was conducted using MEDLINE, CINAHL, and Cochrane Database of Systematic Reviews. The quality of each study was assessed using the Joanna Briggs Critical Appraisal Tools. A total of 16 studies were included. Results: There was evidence that an exclusive HM diet reduces the risk of necrotizing enterocolitis (NEC) for infants with CHD. Evidence with a higher risk for bias indicated that a well-managed HM diet may be associated with improved growth, shorter length of stay, and improved postoperative feeding and nutritional outcomes. Chylothorax outcomes were similar between modified HM and medium-chain triglyceride formula. The studies had significant limitations related to power, lack of control for covariates, and inconsistent delineation of feeding groups. Conclusions: Based on the reduced risk for NEC and given the conclusive benefits in other vulnerable populations, we recommend that clinicians and institutions prioritize programs to support HM feeding for infants with CHD. Large high-quality studies are needed to validate these results. Future work should clarify best practices in managing an HM diet to support optimal growth and development for these infants.
Collapse
Affiliation(s)
| | | | - Erica N Schorr
- University of Minnesota School of Nursing, Minneapolis, Minnesota, USA
| | - Kavisha M Shah
- University of Minnesota School of Nursing, Minneapolis, Minnesota, USA.,Department of Pediatrics, M Health Fairview University of Minnesota Masonic Children's Hospital, Minneapolis, Minnesota, USA
| | - Anna L Trebilcock
- University of Minnesota School of Nursing, Minneapolis, Minnesota, USA
| | - Sara E Ramel
- University of Minnesota School of Nursing, Minneapolis, Minnesota, USA.,Department of Pediatrics, M Health Fairview University of Minnesota Masonic Children's Hospital, Minneapolis, Minnesota, USA
| | - Matthew B Ambrose
- University of Minnesota School of Nursing, Minneapolis, Minnesota, USA.,Department of Pediatrics, M Health Fairview University of Minnesota Masonic Children's Hospital, Minneapolis, Minnesota, USA
| | - Nellie Munn Swanson
- University of Minnesota School of Nursing, Minneapolis, Minnesota, USA.,Department of Nursing, Children's Minnesota, Minneapolis, Minnesota, USA
| | | | - Diane L Spatz
- Department of Family and Community Health, University of Pennsylvania School of Nursing, Philadelphia, Pennsylvania, USA.,Center for Nursing Research & Evidence Based Practice, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
42
|
Kazemian S, Fallahi M, Kazemian M. Reply to the commentary "Liquid gold: do we need to fraction fresh colostrum for oral immunotherapy in premature infants?". Int Breastfeed J 2022; 17:26. [PMID: 35387665 PMCID: PMC8985281 DOI: 10.1186/s13006-022-00473-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 04/03/2022] [Indexed: 11/10/2022] Open
Affiliation(s)
- Sina Kazemian
- Neonatal Health Research Center, Research Institute for Children's Health, Mofid Children's Hospital, Shahid Beheshti University of Medical Sciences, Shariati Ave, Tehran, 15468- 15514, Iran
| | - Minoo Fallahi
- Neonatal Health Research Center, Research Institute for Children's Health, Mofid Children's Hospital, Shahid Beheshti University of Medical Sciences, Shariati Ave, Tehran, 15468- 15514, Iran
| | - Mohammad Kazemian
- Neonatal Health Research Center, Research Institute for Children's Health, Mofid Children's Hospital, Shahid Beheshti University of Medical Sciences, Shariati Ave, Tehran, 15468- 15514, Iran.
| |
Collapse
|
43
|
Salvatori G, De Rose DU, Massolo AC, Patel N, Capolupo I, Giliberti P, Evangelisti M, Parisi P, Toscano A, Dotta A, Di Nardo G. Current Strategies to Optimize Nutrition and Growth in Newborns and Infants with Congenital Heart Disease: A Narrative Review. J Clin Med 2022; 11:1841. [PMID: 35407451 PMCID: PMC8999967 DOI: 10.3390/jcm11071841] [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: 01/29/2022] [Revised: 03/22/2022] [Accepted: 03/24/2022] [Indexed: 11/24/2022] Open
Abstract
(1) Objective: This review aims to identify the clinical and practical barriers to optimizing nutrition in newborn infants with congenital heart disease (CHD) and to describe updated evidence-based recommendations for clinical and nutritional management of these patients in a narrative review. (2) Research Methods and Procedures: We conducted a search of the relevant literature published from 2000 to December 2021. (3) Results: CHD patients undergo several nutritional challenges related to the underlying cardiac disease anomaly, the potential increased risk of NEC, and delayed enteral feeding, resulting in inadequate energy intake and sub-optimal growth, increased morbidity and mortality. (4) Conclusions: To optimize nutrition and growth in newborn infants with CHD, standardized protocols should be implemented. Regular nutritional and growth assessment with a multi-disciplinary team is essential. We propose a decisional algorithm that may represent a potentially useful tool to guide clinicians to optimize growth and nutrition.
Collapse
Affiliation(s)
- Guglielmo Salvatori
- Neonatal Intensive Care Unit, Medical and Surgical Department of Fetus-Newborn-Infant, “Bambino Gesù” Children’s Hospital, IRCCS, 00165 Rome, Italy; (D.U.D.R.); (A.C.M.); (I.C.); (P.G.); (A.D.)
- Human Milk Bank, Medical and Surgical Department of Fetus-Newborn-Infant, “Bambino Gesù” Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Domenico Umberto De Rose
- Neonatal Intensive Care Unit, Medical and Surgical Department of Fetus-Newborn-Infant, “Bambino Gesù” Children’s Hospital, IRCCS, 00165 Rome, Italy; (D.U.D.R.); (A.C.M.); (I.C.); (P.G.); (A.D.)
- PhD Course in Microbiology, Immunology, Infectious Diseases, and Transplants (MIMIT), Faculty of Medicine and Surgery, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Anna Claudia Massolo
- Neonatal Intensive Care Unit, Medical and Surgical Department of Fetus-Newborn-Infant, “Bambino Gesù” Children’s Hospital, IRCCS, 00165 Rome, Italy; (D.U.D.R.); (A.C.M.); (I.C.); (P.G.); (A.D.)
- Human Milk Bank, Medical and Surgical Department of Fetus-Newborn-Infant, “Bambino Gesù” Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Neil Patel
- Department of Neonatology, Royal Hospital for Children, Glasgow G51 4TF, UK;
| | - Irma Capolupo
- Neonatal Intensive Care Unit, Medical and Surgical Department of Fetus-Newborn-Infant, “Bambino Gesù” Children’s Hospital, IRCCS, 00165 Rome, Italy; (D.U.D.R.); (A.C.M.); (I.C.); (P.G.); (A.D.)
- Human Milk Bank, Medical and Surgical Department of Fetus-Newborn-Infant, “Bambino Gesù” Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Paola Giliberti
- Neonatal Intensive Care Unit, Medical and Surgical Department of Fetus-Newborn-Infant, “Bambino Gesù” Children’s Hospital, IRCCS, 00165 Rome, Italy; (D.U.D.R.); (A.C.M.); (I.C.); (P.G.); (A.D.)
- Human Milk Bank, Medical and Surgical Department of Fetus-Newborn-Infant, “Bambino Gesù” Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Melania Evangelisti
- NESMOS Department, Chair of Pediatrics, Faculty of Medicine and Psychology, Sapienza University of Rome, Sant’Andrea University Hospital, 00189 Rome, Italy; (M.E.); (P.P.); (G.D.N.)
| | - Pasquale Parisi
- NESMOS Department, Chair of Pediatrics, Faculty of Medicine and Psychology, Sapienza University of Rome, Sant’Andrea University Hospital, 00189 Rome, Italy; (M.E.); (P.P.); (G.D.N.)
| | - Alessandra Toscano
- Perinatal Cardiology Unit, Medical and Surgical Department of Fetus-Newborn-Infant, “Bambino Gesù” Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Andrea Dotta
- Neonatal Intensive Care Unit, Medical and Surgical Department of Fetus-Newborn-Infant, “Bambino Gesù” Children’s Hospital, IRCCS, 00165 Rome, Italy; (D.U.D.R.); (A.C.M.); (I.C.); (P.G.); (A.D.)
- Human Milk Bank, Medical and Surgical Department of Fetus-Newborn-Infant, “Bambino Gesù” Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Giovanni Di Nardo
- NESMOS Department, Chair of Pediatrics, Faculty of Medicine and Psychology, Sapienza University of Rome, Sant’Andrea University Hospital, 00189 Rome, Italy; (M.E.); (P.P.); (G.D.N.)
| |
Collapse
|
44
|
Tan X, Zhou Y, Xu L, Zhang L, Wang J, Yang W. The predictors of necrotizing enterocolitis in newborns with low birth weight: A retrospective analysis. Medicine (Baltimore) 2022; 101:e28789. [PMID: 35363166 PMCID: PMC9282129 DOI: 10.1097/md.0000000000028789] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/20/2022] [Indexed: 01/04/2023] Open
Abstract
There are needs to investigate the influencing factors of necrotizing enterocolitis (NEC) in low birth weight (LBW) newborns, to provide insights into the clinical management of NEC.This study was a retrospective cohort study. Infants admitted to our hospital from January 1, 2019 to June 30, 2021 were selected. The clinical characteristics of NEC and no-NEC infants were evaluated. Logistic regression analyses were conducted to assess the risk factors of NEC in LBW infants.A total of 192 LBW infants were included, the incidence of NEC in LBW infants was 35.42%. There were significant differences in the congenital heart disease, sepsis, breastfeeding, blood transfusion and probiotics feeding between NEC and no-NEC group (all P < .05), and there were no significant differences in birth weight, gestational age, mother's pregnancy-induced hypertension, premature rupture of fetal membrane, amniotic fluid pollution, fetal asphyxia, neonatal respiratory distress syndrome and mechanical ventilation between NEC and no-NEC group (all P > .05). Congenital heart disease (OR: 2.128, 95% CI: 1.103-3.511), sepsis (OR: 1.630, 95% CI: 1.022-2.549), and blood transfusion (OR: 1.451, 95% CI: 1.014-2.085) were the independent risk factors for NEC in LBW infants, and breastfeeding (OR: 0.494, 95% CI: 0.023-0.928), probiotics feeding (OR: 0.816, 95% CI: 0.782-0.982) were the protective factors for the NEC in LBW infants. The prognosis of NEC infants undergone surgery treatment was better than that of infants undergone conservative treatments (P = .043).The incidence of NEC in LBW is high, which is affected by many factors, and comprehensive interventions targeted on the risk and protective factors should be made to improve the prognosis of LBW infants.
Collapse
Affiliation(s)
- Xuerong Tan
- Department of Neonatology, Ya’an People's Hospital, Ya’an, Sichuan, PR China
- Nursing Department, Ya’an People's Hospital, Ya’an, Sichuan, PR China
| | - Yunxia Zhou
- Department of Neonatology, Ya’an People's Hospital, Ya’an, Sichuan, PR China
| | - Lan Xu
- Department of Neonatology, Ya’an People's Hospital, Ya’an, Sichuan, PR China
| | - Li Zhang
- Department of Neonatology, Ya’an People's Hospital, Ya’an, Sichuan, PR China
| | - Jiaying Wang
- Department of Neonatology, Ya’an People's Hospital, Ya’an, Sichuan, PR China
| | - Wenqiong Yang
- Department of Neonatology, Ya’an People's Hospital, Ya’an, Sichuan, PR China
| |
Collapse
|
45
|
Lund AM, Domellöf M, Pivodic A, Hellström A, Stoltz Sjöström E, Hansen-Pupp I. Mother's Own Milk and Its Relationship to Growth and Morbidity in a Population-based Cohort of Extremely Preterm Infants. J Pediatr Gastroenterol Nutr 2022; 74:292-300. [PMID: 34759238 PMCID: PMC8788942 DOI: 10.1097/mpg.0000000000003352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 10/27/2021] [Indexed: 12/10/2022]
Abstract
OBJECTIVES The aim of the study was to evaluate the relationships between intake of mother's own milk (MOM), compared with intake of pasteurized donor milk (DM), and postnatal growth, incidence of retinopathy of prematurity (ROP) and bronchopulmonary dysplasia (BPD), in extremely preterm infants. METHODS Swedish population-based cohort of surviving extremely preterm infants born 2004 to 2007. Exposure to MOM and DM was investigated from birth until 32 weeks postmenstrual age (PMA) in 453 infants. Primary outcome variables were change in z-score (Δ) from birth to 32 weeks PMA for weight, length, and head circumference (HC). Secondary outcomes were incidence of ROP and BPD. Mixed models adjusting for confounders were used to investigate the association between exposures and outcomes. RESULTS Infants' mean gestational age (GA) was 25.4 weeks. Unadjusted, MOM (per 10 mL · kg-1 · day-1) was associated with Δweight and ΔHC with beta estimates of 0.03 z-score units (95% CI, 0.02-0.04, P < 0.001) and 0.03 z-score units (95% CI, 0.01-0.05, P = 0.003), respectively. After adjustment for predefined confounders, the association remained significant for Δweight and ΔHC. A similar pattern was found between Δweight and each 10% increase of MOM. Unadjusted, a higher intake of MOM (mL · kg-1 · day-1) was significantly associated to a lower probability of any ROP and severe ROP; however, these associations did not remain in the adjusted analyses. No associations were found between MOM (mL · kg-1 · day-1) and BPD. Moreover, no associations were found between DM and growth or morbidity outcomes. CONCLUSIONS An increased intake of MOM, as opposed to DM (and not formula feeding), was associated with improved postnatal weight gain and HC growth from birth until 32 weeks PMA in extremely preterm infants. Interventions aiming at increasing early intake of unpasteurized MOM for extremely preterm infants should be encouraged.
Collapse
Affiliation(s)
- Anna-My Lund
- Division of Paediatrics, Department of Clinical Sciences, Lund, Lund University and Skåne University Hospital
| | - Magnus Domellöf
- Division of Paediatrics, Department of Clinical Sciences, Umeå University
| | - Aldina Pivodic
- Section for Ophthalmology, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg
| | - Ann Hellström
- Section for Ophthalmology, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg
| | | | - Ingrid Hansen-Pupp
- Division of Paediatrics, Department of Clinical Sciences, Lund, Lund University and Skåne University Hospital
| |
Collapse
|
46
|
Reyes SM, Patra B, Elliott MJ. The Impact of Homogenization on Donor Human Milk and Human Milk-Based Fortifiers and Implications for Preterm Infant Health. Curr Dev Nutr 2022; 6:nzab147. [PMID: 35059551 PMCID: PMC8764228 DOI: 10.1093/cdn/nzab147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 12/30/2022] Open
Abstract
An exclusive human milk diet (EHMD) has been shown to reduce health complications of prematurity in infants born weighing ≤1250 g compared with cow milk-based diets. Accordingly, the number of available human milk (HM)-based nutritional products continues to increase. Newly available products, and those reportedly soon to enter the market, include homogenized donor HM and homogenized HM-based fortifiers. Existing literature demonstrating the benefits of an EHMD, however, is limited to non-homogenized HM-based products. Herein, we summarize existing evidence on the impact of homogenization on HM, with a particular focus on changes to the macromolecular structure of the milk fat globule and the subsequent impact on digestion kinetics. We use these published data to create a conceptual framework for the potential implications of homogenized HM-based nutritional products on preterm infant health. Importantly, we underscore that the safety and efficacy of homogenized HM-based products warrant investigation.
Collapse
Affiliation(s)
| | | | - Melinda J Elliott
- Prolacta Bioscience®, Duarte, CA, USA
- Pediatrix Medical Group of Maryland, Rockville, MD, USA
| |
Collapse
|
47
|
Luo P, Zhang K, Chen Y, Geng X, Wu T, Li L, Zhou P, Jiang PP, Ma L. Antenatal Antibiotic Exposure Affects Enteral Feeding, Body Growth, and Neonatal Infection in Preterm Infants: A Retrospective Study. Front Pediatr 2021; 9:750058. [PMID: 35004538 PMCID: PMC8727690 DOI: 10.3389/fped.2021.750058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Antibiotics are widely prescribed by obstetricians, which exposes a large number of infants to antenatal antibiotics (AAB). The effect of AAB on various aspects of neonatal development of preterm infants remains unclear. Methods: In this retrospective study, infants born with gestational age (GA) between 22 +0 and 36 +6 weeks at our unit from 2017 to 2019 were included. Multivariable analysis was adopted to examine the associations between AAB exposure and various outcomes related to enteral feeding process, body growth, and neonatal infection after adjusting for potential confounders. Further subanalysis on the exposure level of AAB and stratified analysis by GA (<34 vs. ≥34 weeks) were also conducted. Results: In this cohort comprising 2,543 preterm infants, AAB was associated with decreased risks of feeding intolerance (odds ratio [OR]: 0.63, 95% confidence interval [CI]: 0.48-0.82) and neonatal infection (OR: 0.63, 95% CI: 0.41-0.94). Higher AAB exposure level was associated with higher Z scores of birth weight (β = 0.37, 95% CI: 0.27-0.47), but lower Δbodyweight Z-scores (β = -0.20, 95% CI: -0.27 to -0.13). AAB was positively associated with the parameters related to body growth in infants with GA <34 weeks but negatively associated in those with GA ≥34 weeks. Conclusions: AAB exposure affects the enteral feeding process and neonatal infection. The effects on body growth vary by the exposure level of AAB and GA of infants. A well-designed prospective and preferably multi-centre study with predefined parameters is required to confirm our findings.
Collapse
Affiliation(s)
- Ping Luo
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Kun Zhang
- Department of Obstetrics, Bao'an Women and Children's Hospital, Jinan University, Shenzhen, China
| | - You Chen
- Department of Neonatology, Bao'an Women and Children's Hospital, Jinan University, Shenzhen, China
| | - Xiuwen Geng
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Tong Wu
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Li Li
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ping Zhou
- Department of Neonatology, Bao'an Women and Children's Hospital, Jinan University, Shenzhen, China
| | - Ping-Ping Jiang
- School of Public Health, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, China
| | - Liya Ma
- Department of Children Healthcare, Bao'an Women and Children's Hospital, Jinan University, Shenzhen, China
| |
Collapse
|
48
|
Percent mother's own milk feedings for preterm neonates predicts discharge feeding outcomes. J Perinatol 2021; 41:2766-2773. [PMID: 34526659 DOI: 10.1038/s41372-021-01205-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 08/09/2021] [Accepted: 09/03/2021] [Indexed: 11/08/2022]
Abstract
INTRODUCTION No studies have determined if there is a threshold whereby use of mother's own milk (MOM) during hospitalization predicts exclusive MOM feeding at discharge. METHODS Among 113 very low birthweight neonates, the ratio of MOM to enteral feeds was measured in the first 14 days, 28 days, and overall hospital stay. The primary outcome was exclusive MOM feeding at discharge. RESULTS For every 1% increase in MOM consumption in the first 14 and 28 days, the odds of being discharge home on an exclusive MOM diet increased nearly 7-fold (OR 7.01, 95% CI: 2.09-23.50) and 17-fold (OR 17.46, 95% CI 4.67-63.31), respectively. A threshold of >50%, >83%, and >85% MOM consumption compared to overall enteral feeds in the first 14 days, 28 days, and throughout hospitalization, respectively, is recommended. CONCLUSIONS Promotion of MOM consumption in the first 2-4 weeks is of paramount importance, with negligible impact of increasing MOM consumption after 28 days.
Collapse
|
49
|
Thermoultrasonication, ultraviolet-C irradiation, and high-pressure processing: Novel techniques to preserve insulin in donor human milk. Clin Nutr 2021; 40:5655-5658. [PMID: 34666256 DOI: 10.1016/j.clnu.2021.09.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/03/2021] [Accepted: 09/16/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND & AIMS Donor human milk (DHM) is recommended as the first alternative for preterm infants if their mother's own milk is not available or if the quantity is not sufficient. The most commonly used technique to eliminate microbial contaminants in DHM is holder pasteurization (HoP). However, the heating process during HoP partially destroys milk bioactive factors such as insulin. Therefore, innovative techniques have been developed as alternatives to HoP. The objective of this study was to determine the effect of HoP, high-temperature-short-time (HTST), thermoultrasonication (TUS), ultraviolet-C irradiation (UV-C), and high-pressure processing (HPP) on the insulin concentration in DHM. METHODS Milk samples from 28 non-diabetic mothers were collected. The milk samples were aliquoted and either left untreated or treated with HoP (62.5 °C; 30 min), HTST (72 °C; 15 s), TUS (60 W; 6 min), UV-C (4863 J/L), or HPP (500 MPa; 5 min). RESULTS The mean insulin concentration in untreated milk was 79 ± 41 pmol/L. The mean insulin retention rate was 67% for HoP, 78% for HTST, 97% for TUS, 94% for UV-C, and 106% for HPP. The mean insulin concentration in milk treated with HoP was significantly lower compared to untreated milk (p = 0.01). CONCLUSION TUS, UV-C, and HPP preserve insulin in DHM. The insulin concentration in DHM is affected to a larger extent by HoP than by HTST. These results indicate that TUS, UV-C, and HPP may serve as alternatives to HoP.
Collapse
|
50
|
Abstract
Microbes in the 21st century are understood as symbionts ‘completing’ the human ‘superorganism’ (Homo sapiens plus microbial partners-in-health). This paper addresses a significant paradox: despite the vast majority of our genes being microbial, the lack of routine safety testing for the microbiome has led to unintended collateral side effects from pharmaceuticals that can damage the microbiome and inhibit innate ‘colonization resistance’ against pathobionts. Examples are discussed in which a Microbiome First Medicine approach provides opportunities to ‘manage our microbes’ holistically, repair dysbiotic superorganisms, and restore health and resilience in the gut and throughout the body: namely, managing nosocomial infections for Clostridioides difficile and Staphylococcus aureus and managing the gut and neural systems (gut–brain axis) in autism spectrum disorder. We then introduce a risk analysis tool: the evidence map. This ‘mapping’ tool was recently applied by us to evaluate evidence for benefits, risks, and uncertainties pertaining to the breastmilk ecosystem. Here, we discuss the potential role of the evidence map as a risk analysis methodology to guide scientific and societal efforts to: (1) enhance ecosystem resilience, (2) ‘manage our microbes’, and (3) minimize the adverse effects of both acute and chronic diseases.
Collapse
|