1
|
Abdullahi AM, Zhao S, Xu Y. Efficacy of probiotic supplementation in preventing necrotizing enterocolitis in preterm infants: a systematic review and meta-analysis. J Matern Fetal Neonatal Med 2025; 38:2485215. [PMID: 40204632 DOI: 10.1080/14767058.2025.2485215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 02/27/2025] [Accepted: 03/22/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a severe condition in preterm infants, involving intestinal inflammation and bacterial invasion, leading to high morbidity and mortality. Probiotics may reduce NEC by promoting beneficial gut bacteria, but the role of Bifidobacterium bifidum G001 (BBG001) is not well understood. This meta-analysis evaluates the effectiveness of BBG001 versus placebo in preventing NEC, late-onset sepsis, and all-cause mortality in preterm infants. METHODS Following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we conducted a systematic review and meta-analysis of randomized controlled trials (RCTs) from 2014 to 2024. Databases searched included PubMed, Cochrane Central Register, MEDLINE, and EMBASE. Inclusion criteria encompassed RCTs involving preterm infants (<37 weeks gestation and/or <2500 g birth weight) receiving enteral probiotics, including BBG001, compared to placebo. Outcomes assessed were incidence of NEC (≥stage II), late-onset sepsis, and all-cause mortality. Data extraction and quality assessment were independently performed. RESULTS From 430 identified records, nine studies involving 7180 preterm infants met the inclusion criteria. BBG001 significantly reduced the risk of NEC stage >2 (OR = 0.66; 95% CI [0.47, 0.94], p = .02), mortality and morbidity (OR = 0.74; 95% CI [0.62, 0.89], p = .001), and sepsis (OR = 0.71; 95% CI [0.53, 0.93], p = .01). It also lowered the odds of periventricular leukomalacia (OR = 0.61; 95% CI [0.47, 0.78], p < .0001) and intraventricular hemorrhage (OR = 0.48; 95% CI [0.33, 0.69], p < .0001). Additionally, BBG001 showed a protective effect against infection-related outcomes, reducing the odds of death attributed to infection (OR = 0.80; 95% CI: 0.65, 0.99; p = .04). CONCLUSIONS Probiotic supplementation with BBG001 appears effective in reducing NEC incidence, bloodstream infections, and infection-related deaths in preterm infants. Despite promising results, variability in study designs necessitates further large-scale RCTs to confirm these findings and establish BBG001's role in neonatal care.
Collapse
Affiliation(s)
| | - Shuangkui Zhao
- Department of NICU, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanping Xu
- Department of NICU, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
2
|
Zhang L, Ma Y, Hettinga K, Zhou P. Suckling Rat Pup Model: Do Caprine Milk Lactoferrin and Immunoglobulins Have Different Digestion and Absorption Properties from That of Human and Bovine Species? JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:3069-3079. [PMID: 39873219 DOI: 10.1021/acs.jafc.4c10539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
This study aimed to investigate the digestion and absorption properties of caprine milk serum proteins in comparison to human and bovine species by using rat pups to mimic preterm infants. The results indicate that caprine lactoferrin (LTF) had a shorter retention time in the intestine and released a greater number of fragments, resembling human milk LTF more closely. In contrast, caprine immunoglobulins (Igs) were similar to bovine Igs and both exhibited a longer retention time in the intestine. For absorption, caprine Igs could be absorbed intact, which was similar to human and bovine Igs, whereas caprine LTF fragments were found in jejunum but not in plasma of rat pups. This is similar to bovine LTF but differed from human LTF as human LTF could be absorbed intact in plasma of rat pups at 20 min. In addition, the absorption rate of peptides and amino acids from caprine milk serum was similar to that of human milk serum, which was higher than that from bovine milk serum. This study aimed to enhance our understanding of the differences in bioavailability of LTF and Igs derived from caprine, human milk, and bovine milk, thereby offering guidance for selecting protein sources for premature infants.
Collapse
Affiliation(s)
- Lina Zhang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Laboratory for Dairy Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ying Ma
- National R&D Center for Se-rich Agricultural Products Processing, Hubei Engineering Research Center for Deep Processing of Green Se-rich Agricultural Products, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Kasper Hettinga
- International Joint Research Laboratory for Dairy Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- Food Quality & Design Group, Wageningen University, Wageningen 6708WG, The Netherlands
| | - Peng Zhou
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Laboratory for Dairy Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
3
|
Brown JA, Bashir H, Zeng MY. Lifelong partners: Gut microbiota-immune cell interactions from infancy to old age. Mucosal Immunol 2025:S1933-0219(25)00006-6. [PMID: 39862964 DOI: 10.1016/j.mucimm.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
Our immune system and gut microbiota are intricately coupled from birth, both going through maturation during early life and senescence during aging almost in a synchronized fashion. The symbiotic relationship between the human host and microbiota is critically dependent on a healthy immune system to keep our microbiota in check, while the microbiota provides essential functions to promote the development and fitness of our immune system. The partnership between our immune system and microbiota is particularly important during early life, when microbial ligands and metabolites shape the development of the immune cells and immune tolerance; during aging, having sufficient beneficial gut bacteria is critical for the maintenance of intact mucosal barriers, immune metabolic fitness, and strong immunity against pathogens. The immune system during childhood is programmed, with the support of the microbiota, to develop robust immune tolerance, and limit autoimmunity and metabolic dysregulation, which are prevalent during aging. This review comprehensively explores the mechanistic underpinnings of gut microbiota-immune cell interactions during infancy and old age, with the goal to gain a better understanding of potential strategies to leverage the gut microbiota to combat age-related immune decline.
Collapse
Affiliation(s)
- Julia A Brown
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States
| | - Hilal Bashir
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States
| | - Melody Y Zeng
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School, New York, NY 10065, United States.
| |
Collapse
|
4
|
Marissen J, Reichert L, Härtel C, Fortmann MI, Faust K, Msanga D, Harder J, Zemlin M, Gomez de Agüero M, Masjosthusmann K, Humberg A. Antimicrobial Peptides (AMPs) and the Microbiome in Preterm Infants: Consequences and Opportunities for Future Therapeutics. Int J Mol Sci 2024; 25:6684. [PMID: 38928389 PMCID: PMC11203687 DOI: 10.3390/ijms25126684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Antimicrobial peptides (AMPs) are crucial components of the innate immune system in various organisms, including humans. Beyond their direct antimicrobial effects, AMPs play essential roles in various physiological processes. They induce angiogenesis, promote wound healing, modulate immune responses, and serve as chemoattractants for immune cells. AMPs regulate the microbiome and combat microbial infections on the skin, lungs, and gastrointestinal tract. Produced in response to microbial signals, AMPs help maintain a balanced microbial community and provide a first line of defense against infection. In preterm infants, alterations in microbiome composition have been linked to various health outcomes, including sepsis, necrotizing enterocolitis, atopic dermatitis, and respiratory infections. Dysbiosis, or an imbalance in the microbiome, can alter AMP profiles and potentially lead to inflammation-mediated diseases such as chronic lung disease and obesity. In the following review, we summarize what is known about the vital role of AMPs as multifunctional peptides in protecting newborn infants against infections and modulating the microbiome and immune response. Understanding their roles in preterm infants and high-risk populations offers the potential for innovative approaches to disease prevention and treatment.
Collapse
Affiliation(s)
- Janina Marissen
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
- Würzburg Institute of Systems Immunology, Max-Planck Research Group, University of Würzburg, 97078 Würzburg, Germany;
| | - Lilith Reichert
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
| | - Christoph Härtel
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
- German Center for Infection Research, Site Hamburg-Lübeck-Borstel-Riems, 23538 Lübeck, Germany
| | - Mats Ingmar Fortmann
- Department of Pediatrics, University Hospital Schleswig-Holstein, 23538 Lübeck, Germany; (M.I.F.); (K.F.)
| | - Kirstin Faust
- Department of Pediatrics, University Hospital Schleswig-Holstein, 23538 Lübeck, Germany; (M.I.F.); (K.F.)
| | - Delfina Msanga
- Department of Pediatrics, Bugando Hospital, Catholic University of Health and Allied Sciences, Mwanza 33109, Tanzania;
| | - Jürgen Harder
- Department of Dermatology, Venerology and Allergology, Quincke Research Center, Kiel University, 24105 Kiel, Germany;
| | - Michael Zemlin
- Department of General Pediatrics and Neonatology, Saarland University Medical Center, 66421 Homburg, Germany;
| | - Mercedes Gomez de Agüero
- Würzburg Institute of Systems Immunology, Max-Planck Research Group, University of Würzburg, 97078 Würzburg, Germany;
| | - Katja Masjosthusmann
- Department of General Pediatrics, University Children’s Hospital Münster, 48149 Münster, Germany; (K.M.); (A.H.)
| | - Alexander Humberg
- Department of General Pediatrics, University Children’s Hospital Münster, 48149 Münster, Germany; (K.M.); (A.H.)
| |
Collapse
|
5
|
Vardar G, Rzayev T, Tezel KG, Ozek E. Can We Estimate Late-Onset Sepsis by Serial Methemoglobin Levels? An Observational Study in Preterm Neonates. Fetal Pediatr Pathol 2023; 42:753-765. [PMID: 37318102 DOI: 10.1080/15513815.2023.2223308] [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] [Received: 03/21/2023] [Accepted: 06/05/2023] [Indexed: 06/16/2023]
Abstract
Objective: To assess serial methemoglobin (MetHb) levels in preterm infants as a possible diagnostic method for late-onset sepsis (LOS). Methods: Preterm infants were assigned into two groups: those with culture-proven LOS and controls. Serial MetHb levels were measured. Results: The MetHb values of the LOS group were found to be significantly increased (p < 0.001). The cutoff value for the detection of LOS was calculated as MetHb > 1.75%, optimized for a sensitivity of 81.9% and specificity of 90%. After antimicrobial therapy, MetHb values were found to decrease significantly (p < 0.001). MetHb had an AUC of 0.810 for mortality using the calculated cutoff of >2% (p < 0.005). Conclusions: MetHb levels increase at the onset of LOS and decrease following treatment. MetHb can be added to other sepsis biomarkers as a rapid infectious process indicator for preterm neonates. MetHb > 2% is associated with LOS mortality.
Collapse
Affiliation(s)
- Gonca Vardar
- Division of Neonatology, Department of Pediatrics, School of Medicine, Marmara University, Pendik/Istanbul, Turkey
| | - Turkay Rzayev
- Division of Neonatology, Department of Pediatrics, School of Medicine, Marmara University, Pendik/Istanbul, Turkey
| | - Kubra Gokce Tezel
- Division of Neonatology, Department of Pediatrics, School of Medicine, Marmara University, Pendik/Istanbul, Turkey
| | - Eren Ozek
- Division of Neonatology, Department of Pediatrics, School of Medicine, Marmara University, Pendik/Istanbul, Turkey
| |
Collapse
|
6
|
Xiang Q, Yan X, Shi W, Li H, Zhou K. Early gut microbiota intervention in premature infants: Application perspectives. J Adv Res 2023; 51:59-72. [PMID: 36372205 PMCID: PMC10491976 DOI: 10.1016/j.jare.2022.11.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/30/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Preterm birth is the leading cause of death in children under the age of five. One of the major factors contributing to the high risk of diseases and deaths in premature infants is the incomplete development of the intestinal immune system. The gut microbiota has been widely recognized as a critical factor in promoting the development and function of the intestinal immune system after birth. However, the gut microbiota of premature infants is at high risk of dysbiosis, which is highly associated with adverse effects on the development and education of the early life immune system. Early intervention can modulate the colonization and development of gut microbiota and has a long-term influence on the development of the intestinal immune system. AIM OF REVIEW This review aims to summarize the characterization, interconnection, and underlying mechanism of gut microbiota and intestinal innate immunity in premature infants, and to discuss the status, applicability, safety, and prospects of different intervention strategies in premature infants, thus providing an overview and outlook of the current applications and remaining gaps of early intervention strategies in premature infants. KEY SCIENTIFIC CONCEPTS OF REVIEW This review is focused on three key concepts. Firstly, the gut microbiota of premature infants is at high risk of dysbiosis, resulting in dysfunctional intestinal immune system processes. Secondly, contributing roles of early intervention have been observed in improving the intestinal environment and promoting gut microbiota colonization, which is significant in the development and function of gut immunity in premature infants. Thirdly, different strategies of early intervention, such as probiotics, fecal microbiota transplantation, and nutrients, show different safety, applicability, and outcome in premature infants, and the underlying mechanism is complex and poorly understood.
Collapse
Affiliation(s)
- Quanhang Xiang
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Xudong Yan
- Department of Neonatal Intensive Care Unit, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Wei Shi
- Department of Obstetrics and Gynecology, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Huiping Li
- Department of Respiratory and Critical Care Medicine, the first affiliated hospital of Southern University of Science and Technology of China, Shenzhen People's Hospital, Shenzhen, China; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| | - Kai Zhou
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
7
|
Miranda-Rius J, Brunet-Llobet L, Blanc V, Álvarez G, Moncunill-Mira J, Mashala EI, Kasebele Y, Masenga G, Nadal A, León R. Microbial profile of placentas from Tanzanian mothers with adverse pregnancy outcomes and periodontitis. Oral Dis 2023; 29:772-785. [PMID: 34255399 DOI: 10.1111/odi.13962] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/04/2021] [Accepted: 07/09/2021] [Indexed: 02/05/2023]
Abstract
AIM To investigate microbial profiles in placentas from a population of East African mothers with and without adverse pregnancy outcomes and with regard to their periodontal status. MATERIAL AND METHODS Thirty-six placentas from pregnant women from Tanzania were classified into three groups according to both pregnancy outcome and the mother's periodontal health. The microbial composition in each group was then compared using 16S rRNA metagenomics. Additionally, placenta specimens were analyzed histologically for chorioamnionitis by a single pathologist blinded to the clinical data. RESULTS The greatest differences were observed in the group of mothers with periodontitis. The microbial load was low in all three groups of mothers. Periodontitis had a notable influence on the structure of the placental microbiota. Three phyla and 44 genera were associated with periodontitis, whereas only the Tenericutes phylum was associated with the adverse pregnancy variable. Streptococcaceae and Mycoplasmataceae families were associated with both periodontitis and adverse pregnancy outcomes. Finally, although the differences for chorioamnionitis were not significant, this intra-amniotic infection was more frequent in the placentas from mothers with periodontitis. CONCLUSIONS Our findings suggest that bacteria from the oral cavity may involve the feto-placental unit, and that periodontitis may be a modulating factor of the microbial community present in this niche.
Collapse
Affiliation(s)
- Jaume Miranda-Rius
- Department of Odontostomatology, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,Department of Dentistry, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain.,Hospital Dentistry, Clinical Orthodontics and Periodontal Medicine Research Group (HDCORPEMrg), Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| | - Lluís Brunet-Llobet
- Department of Dentistry, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain.,Hospital Dentistry, Clinical Orthodontics and Periodontal Medicine Research Group (HDCORPEMrg), Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| | - Vanessa Blanc
- Department of Oral Microbiology, Dentaid Research Center, Cerdanyola del Vallès, Barcelona, Spain
| | - Gerard Álvarez
- Department of Oral Microbiology, Dentaid Research Center, Cerdanyola del Vallès, Barcelona, Spain
| | - Jordi Moncunill-Mira
- Department of Dentistry, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain.,Hospital Dentistry, Clinical Orthodontics and Periodontal Medicine Research Group (HDCORPEMrg), Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| | - Elias I Mashala
- Scholarship Programme for Young African Researchers, Doctoral Programme in Medicine and Translational Research, Faculty of Medicine and Health Sciences, The Coimbra Group of Universities, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | - Yona Kasebele
- Department of Pathology, Kilimanjaro Christian Medical Centre (KCMC), Kilimanjaro Christian Medical University College (KCMUCo, Moshi, Tanzania
| | - Gileard Masenga
- Department of Obstetrics and Gynecology, Kilimanjaro Christian Medical Centre (KCMC), Kilimanjaro Christian Medical University College (KCMUCo, Moshi, Tanzania
| | - Alfons Nadal
- Department of Pathology, Hospital Clínic, University of Barcelona, August Pi i Sunyer Biomedical Research Institute (IDIBAPS, Barcelona, Spain
| | - Rubén León
- Department of Oral Microbiology, Dentaid Research Center, Cerdanyola del Vallès, Barcelona, Spain
| |
Collapse
|
8
|
Salaria N, Neeraj, Furhan J, Kumar R. Gut Microbiome: Perspectives and Challenges in Human Health. ROLE OF MICROBES IN SUSTAINABLE DEVELOPMENT 2023:65-87. [DOI: 10.1007/978-981-99-3126-2_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
9
|
Qi Q, Wang L, Gebremedhin MA, Li S, Wang X, Shen J, Zhu Y, Andegiorgish AK, Cheng Y, Shi L, Zhou J, Yang Y, Kang Y, Yang W, Zhu Z, Zeng L. The impact of early-life antibiotics and probiotics on gut microbial ecology and infant health outcomes: a Pregnancy and Birth Cohort in Northwest China (PBCC) study protocol. BMC Pediatr 2022; 22:738. [PMID: 36577993 PMCID: PMC9795439 DOI: 10.1186/s12887-022-03811-3] [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: 12/22/2021] [Accepted: 12/18/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Unreasonable use of antibiotics and probiotics can alter the gut ecology, leading to antibiotic resistance and suboptimal health outcomes during early life. Our study aims are to clarify the association among antibiotic and probiotic exposure in early life, the microecology of the gut microbiota, and the development of antibiotic resistance; to investigate the long-term impact of antibiotics and probiotics on the health outcomes of infants and young children; and to provide a theoretical basis for the rational use of antibiotics and probiotics from a life course perspective. METHODS The study is a prospective, longitudinal birth cohort study conducted in Shaanxi Province, China from 2018 to 2024. A total of 3,000 eligible mother-child pairs will be enrolled from rural, suburban, and urban areas. The recruitment of the participants begins at pregnancy, and the newborns will be followed up for 2 years at successive timepoints: within 3 days after birth, 42 days after birth, and at 3, 6, 12, 18, and 24 months of age. Sociodemographic data, environmental exposures, dietary patterns, psychological conditions, and medical and drug histories are collected. Cognitive and behavioural development among infants and young children and questionnaires on antibiotic knowledge and behaviour among caregivers will be collected at 12 and 24 months of age. The faecal samples are collected and analysed by 16S rRNA high-throughput sequencing and quantitative PCR (qPCR) for antibiotic resistance genes. DISCUSSION The findings will inform antibiotic and probiotic use for pregnant women and infants and contribute to establishing rational use strategies of antibiotics and probiotics for paediatricians, health practitioners, and drug administration policy-makers. TRIAL REGISTRATION The study was registered on the Chinese Clinical Trial Registry (ChiCTR) platform, http://www.chictr.org.cn (Record ID: ChiCTR2100047531, June 20, 2021).
Collapse
Affiliation(s)
- Qi Qi
- School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi, People's Republic of China
| | - Liang Wang
- School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi, People's Republic of China
| | - Mitslal Abrha Gebremedhin
- School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi, People's Republic of China
| | - Shaoru Li
- School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi, People's Republic of China
| | - Xueyao Wang
- School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi, People's Republic of China
| | - Jiali Shen
- School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi, People's Republic of China
| | - Yingze Zhu
- School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi, People's Republic of China
| | - Amanuel Kidane Andegiorgish
- School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi, People's Republic of China
| | - Yue Cheng
- School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi, People's Republic of China
| | - Lu Shi
- School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi, People's Republic of China
| | - Jing Zhou
- Department of Paediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Ying Yang
- Xi'an Lintong District Maternal and Child Health Hospital, Xi'an, Shaanxi, People's Republic of China
| | - Yijun Kang
- School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi, People's Republic of China
| | - Wenfang Yang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Xi'an Jiaotong University, Maternal & Child Health Centre, Xi'an, Shaanxi, People's Republic of China
| | - Zhonghai Zhu
- School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi, People's Republic of China
| | - Lingxia Zeng
- School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi, People's Republic of China.
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi, People's Republic of China.
| |
Collapse
|
10
|
Rodrigues Wilde MO, Mezadri T, Gouveia PB, Grillo LP, Valete C. Prediction of early-onset neonatal sepsis in umbilical cord blood analysis: an integrative review. J Matern Fetal Neonatal Med 2022; 35:10187-10198. [PMID: 36100266 DOI: 10.1080/14767058.2022.2122798] [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: 10/14/2022]
Abstract
OBJECTIVE The aim of this study was to describe the inflammatory markers studied in umbilical cord blood and to analyze the performance of the three markers most frequently studied for the prediction of early-onset neonatal sepsis. DATA SOURCES An integrative review from 1995 to 2021 was performed, with a search in the MEDLINE, Embase, Cochrane Library, SciELO, and gray literature databases, using the terms "neonates," "newborns," "neonatal sepsis," "early-onset neonatal sepsis," "neonatal infection," "inflammatory markers," "biomarkers," "cord blood," "fetal blood." STUDY SELECTION AND DATA EXTRACTIONS Study evaluation was limited to primary studies, prospective, observational or intervention, descriptive or analytical, that assessed the diagnosis of early-onset neonatal sepsis using inflammatory markers in umbilical cord blood, in Portuguese, English, or Spanish. Qualitative studies, reports, review studies, and case series were excluded. Only studies with a punctuation ≥ 6 in the Newcastle-Ottawa scale were included. RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE Sixteen studies were included in the qualitative synthesis. Procalcitonin, C-reactive protein, and interleukin-6 were the most frequently studied markers. The best performance for C-reactive protein was observed at a 0.2 mg/L cutoff, with a sensitivity of 82% and a negative predictive value of 99%. Procalcitonin presented the best performance at a 0.5 ng/mL cutoff with 87.5% sensitivity and 98.7% negative predictive value. Interleukin-6 presented the best performance at a 108.5 ng/mL cutoff, with 95% sensitivity and 97.4% negative predictive value. CONCLUSION The evaluation of markers in the umbilical cord for the diagnosis of early-onset neonatal sepsis, could contribute to a more assertive therapy for the neonate and anticipate sepsis screening. Since the cost is less and technically easier, C-reactive protein is recommended for routine use.
Collapse
Affiliation(s)
| | - Tatiana Mezadri
- Professional Master's Program in Health and Work Management, University of the Vale do Itajaí, Itajai, Brazil
| | | | - Luciane Peter Grillo
- Professional Master's Program in Health and Work Management, University of the Vale do Itajaí, Itajai, Brazil
| | - Cristina Valete
- Department of Medicine, Universidade Federal de São Carlos, Sao Carlos, Brazil
| |
Collapse
|
11
|
Reyes-García DV, Canul-Euan AA, Rivera-Rueda MA, Cruz-Alvarado CE, Bermejo-Martínez LB, Arreola-Ramírez G, Cordero-González G, Carrera-Muiños S, Diaz-Valencia JD, Estrada-Gutiérrez G, Irles C, Gonzalez-Perez G. Neonatal Antibiotic Treatment Can Affect Stool Pattern and Oral Tolerance in Preterm Infants. Life (Basel) 2022; 12:life12071043. [PMID: 35888130 PMCID: PMC9319374 DOI: 10.3390/life12071043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 11/16/2022] Open
Abstract
Preterm neonates are at high risk of infectious and inflammatory diseases which require antibiotic treatment. Antibiotics influence neonatal gut microbiome development, and intestinal dysbiosis has been associated with delayed gastrointestinal transit. Neonates who take less time to pass meconium have a better tolerance to enteral feeding. We analyzed the effect of neonatal antibiotic treatment on the stool pattern and oral tolerance in 106 preterm infants < 33 weeks gestational age. Neonates were classified in 3 groups according to neonatal antibiotic (ABT) treatment days: no antibiotics, 3−7 d ABT, and ≥8 d ABT. Preterm infants from the ≥8 d ABT group took longer to pass meconium and to start green and yellow stools, took longer to reach 100 and 150 mL/kg/day, and reached reduced volumes in enteral feeds at day of life 14 and 28 than infants from no ABT and 3−7 d ABT groups. Multiple linear regression models showed that neonatal antibiotic treatment, birth weight, invasive mechanical ventilation, surfactant, enteral feeding start day, neonatal parenteral nutrition, and neonatal fasting days are associated with the stool pattern and oral tolerance in preterm infants.
Collapse
Affiliation(s)
- Diana Verónica Reyes-García
- Neonatal Intensive Care Unit, National Institute of Perinatology “Isidro Espinosa de los Reyes”, Mexico City 11000, Mexico; (D.V.R.-G.); (M.A.R.-R.); (C.E.C.-A.); (G.C.-G.); (S.C.-M.)
| | - Arturo Alejandro Canul-Euan
- Department of Developmental Neurobiology, National Institute of Perinatology “Isidro Espinosa de los Reyes”, Mexico City 11000, Mexico;
| | - María Antonieta Rivera-Rueda
- Neonatal Intensive Care Unit, National Institute of Perinatology “Isidro Espinosa de los Reyes”, Mexico City 11000, Mexico; (D.V.R.-G.); (M.A.R.-R.); (C.E.C.-A.); (G.C.-G.); (S.C.-M.)
| | - Claudia Edith Cruz-Alvarado
- Neonatal Intensive Care Unit, National Institute of Perinatology “Isidro Espinosa de los Reyes”, Mexico City 11000, Mexico; (D.V.R.-G.); (M.A.R.-R.); (C.E.C.-A.); (G.C.-G.); (S.C.-M.)
| | - Luisa Bertha Bermejo-Martínez
- Department of Immunobiochemistry, National Institute of Perinatology “Isidro Espinosa de los Reyes”, Mexico City 11000, Mexico;
| | - Gabriela Arreola-Ramírez
- Department of Pediatric Follow-Up, National Institute of Perinatology “Isidro Espinosa de los Reyes”, Mexico City 11000, Mexico;
| | - Guadalupe Cordero-González
- Neonatal Intensive Care Unit, National Institute of Perinatology “Isidro Espinosa de los Reyes”, Mexico City 11000, Mexico; (D.V.R.-G.); (M.A.R.-R.); (C.E.C.-A.); (G.C.-G.); (S.C.-M.)
| | - Sandra Carrera-Muiños
- Neonatal Intensive Care Unit, National Institute of Perinatology “Isidro Espinosa de los Reyes”, Mexico City 11000, Mexico; (D.V.R.-G.); (M.A.R.-R.); (C.E.C.-A.); (G.C.-G.); (S.C.-M.)
| | - Juan Daniel Diaz-Valencia
- Department of Physiology and Cellular Development, National Institute of Perinatology “Isidro Espinosa de los Reyes”, Mexico City 11000, Mexico; (J.D.D.-V.); (C.I.)
| | - Guadalupe Estrada-Gutiérrez
- Research Direction, National Institute of Perinatology “Isidro Espinosa de los Reyes”, Mexico City 11000, Mexico;
| | - Claudine Irles
- Department of Physiology and Cellular Development, National Institute of Perinatology “Isidro Espinosa de los Reyes”, Mexico City 11000, Mexico; (J.D.D.-V.); (C.I.)
| | - Gabriela Gonzalez-Perez
- Department of Physiology and Cellular Development, National Institute of Perinatology “Isidro Espinosa de los Reyes”, Mexico City 11000, Mexico; (J.D.D.-V.); (C.I.)
- Correspondence: ; Tel.: +52-55-55209900 (ext. 340)
| |
Collapse
|
12
|
Zhang Y, Wang O, Mi H, Yi J, Cai S. Rhus chinensis Mill. fruits prevent necrotizing enterocolitis in rat pups via regulating the expressions of key proteins involved in multiple signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2022; 290:115103. [PMID: 35157955 DOI: 10.1016/j.jep.2022.115103] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/03/2022] [Accepted: 02/09/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Based on ancient records and previous studies, many parts of Rhus chinensis Mill., including the fruits, have good preventive and therapeutic effects on inflammation, malaria, diarrhea, and gastrointestinal diseases. Rhus plants and Galla chinensis produced from R. chinensis leaves can also prevent or cure intestinal diseases. However, the preventive effect and molecular mechanisms of R. chinensis fruits on necrotizing enterocolitis (NEC) have not been comprehensively studied. AIM OF THE STUDY This article aims to estimate the effect of the 80% ethanol extract of R. chinensis fruits (RM) on alleviating NEC in rat pups and illustrate the potential molecular mechanisms. MATERIALS AND METHODS Rat pups were subjected to formula feeding, intermittent hypoxic, and cold stresses to establish the NEC model. The preventive effects of RM on NEC were evaluated through survival rate; clinical sickness index; macroscopic conditions; histopathology; and expression levels of inflammatory markers (i.e., tumor necrosis factor-α [TNF-α], interleukin-6 [IL-6]), oxidative stress indicators (i.e., total antioxidant status [TAS], total oxidant status [TOS], superoxide dismutase [SOD], glutathione peroxidase [GSH-Px], myeloperoxidase [MPO], malondialdehyde [MDA]), and tight junction proteins (i.e., Zonula Occludens 1 [ZO-1], Occludin). Moreover, the expression levels of several key proteins involved in oxidative stress (i.e., nuclear factor erythroid 2-related factor 2 [Nrf2], NAD(P)H-quinone oxidoreductase-1 [NQO1]), inflammation (i.e., Toll-like receptor 4 [TLR4], phosphorylated-nuclear factor kappa-B [p-NF-κB], inducible nitric oxide synthase [iNOS]), and apoptosis (i.e., cleaved cysteinyl aspartate specific proteinase-3 [cleaved Caspase-3], Bcl-2-associated X [Bax], B-cell lymphoma-2 [Bcl-2]) in intestinal tissues were analyzed to clarify the molecular mechanisms. RESULTS The extract particularly high doses (400 mg RM/kg body weight) could remarkably reduce the mortality and clinical sickness score and improve the macroscopic condition and histopathological injury of the intestine in NEC pups. After RM administration, the levels of TOS, TNF-α, IL-6, MPO, and MDA in the bowel tissue decreased, whereas the levels of TAS, SOD, and GSH-Px were significantly enhanced. The expression levels of ZO-1 and Occludin proteins were dramatically augmented in RM-treated groups to maintain intestinal barrier integrity. Further analyses revealed that RM might prevent NEC pups by improving some pivotal proteins involved in oxidative stress, inflammation, and apoptosis of enterocytes, namely, by down-regulating the levels of TLR4, p-NF-κB, iNOS, cleaved Caspase-3, and Bax and up-regulating the levels of Bcl-2, NQO1, and Nrf2. CONCLUSIONS The RM prevented the intestinal inflammation and damage caused by NEC by regulating the expression of several pivotal proteins involved in oxidative stress, inflammation, and apoptosis. This study might provide a scientific basis for R. chinensis fruits as a traditional herbal medicine to prevent and/or alleviate NEC.
Collapse
Affiliation(s)
- Yi Zhang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, Yunnan Province, People's Republic of China
| | - Ou Wang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, 100050, People's Republic of China
| | - Hongying Mi
- The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, Yunnan Province, People's Republic of China
| | - Junjie Yi
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, Yunnan Province, People's Republic of China
| | - Shengbao Cai
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, Yunnan Province, People's Republic of China.
| |
Collapse
|
13
|
A Pilot Study To Establish an In Vitro Model To Study Premature Intestinal Epithelium and Gut Microbiota Interactions. mSphere 2021; 6:e0080621. [PMID: 34643422 PMCID: PMC8513685 DOI: 10.1128/msphere.00806-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Intestinal microbiota has emerged as an important player in the health and disease of preterm infants. The interactions between intestinal flora and epithelium can lead to local injury and systemic diseases. A suitable in vitro cell model is needed to enhance our understanding of these interactions. In this study, we exposed fetal epithelial cell cultures (FHs-74 int cells, human, ATCC CCL 241) to sterile fecal filtrates derived from stool collected from preterm infants at <2 and at 3 to 4 weeks of age. We measured the cytokine levels from the culture media after 4, 24, and 48 h of exposure to the fecal filtrates. We analyzed the 16S rRNA V4 gene data of the fecal samples and transcriptome sequencing (RNA-seq) data from the fetal epithelial cells after 48 h of exposure to the same fecal filtrates. The results showed correlations between inflammatory responses (both cytokine levels and gene expression) and the Proteobacteria-to-Firmicutes ratio and between fecal bacterial genera and epithelial apoptosis-related genes. Our in vitro cell model can be further developed and applied to study how the epithelium responds to different microbial flora from preterm infants. Combining immature epithelial cells and preterm infant stool samples into one model allows us to investigate disease processes in preterm infants in a way that had not been previously reported. IMPORTANCE The gut bacterial flora influences the development of the immune system and long-term health outcomes in preterm infants. Studies of the mechanistic interactions between the gut bacteria and mucosal barrier are limited to clinical observations, animal models, and in vitro cell culture models for this vulnerable population. Most in vitro cell culture models of microbe-host interactions use single organisms or adult origin cell lines. Our study is innovative and significant in that we expose immature epithelial cells derived from fetal tissues to fecal filtrates from eight stool samples from four preterm infants to study the role of intestinal epithelial cells. In addition, we analyzed epithelial gene expression to examine multiple cellular processes simultaneously. This model can be developed into patient-derived two- or three-dimensional cell cultures exposed to their own fecal material to allow better prediction of patient physiological responses to support the growing field of precision medicine.
Collapse
|
14
|
Hanford J, Mannebach K, Ohler A, Patten M, Pardalos J. Rates of Comorbidities in Very Low Birth Weight Infants Fed an Exclusive Human Milk Diet Versus a Bovine Supplemented Diet. Breastfeed Med 2021; 16:814-820. [PMID: 34415775 DOI: 10.1089/bfm.2020.0345] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Background: Our level III neonatal intensive care unit (NICU) implemented the use of an exclusive human milk diet (EHD) and sought to determine its effect on the severe co-morbidities of preterm infants as well as the potential cost-savings due to the anticipated reduction in these co-morbidities. Methods: A retrospective cohort study was completed to determine if an EHD statistically decreased the rate of co-morbidities including length of stay (LOS), days on total parental nutrition (TPN), rates of late onset sepsis, necrotizing enterocolitis (NEC), bronchopulmonary dysplasia (BPD), and severe retinopathy of prematurity (ROP). Results: An EHD significantly decreased the odds of severe ROP (adjusted odds-ratio (aOR)=0.349; 95%CI [0.156, 0.739]; p=0.008) and late onset sepsis (aOR=0.323; 95%CI [0.123, 0.768]; p=0.014). Analysis of cost-effectiveness of an EHD relative to a BSD based on the incremental costs of these co-morbidities determined the net loss in direct hospital costs per patient were estimated to be $420 in 2016 US dollars; however, given the long-term health-care costs and non-pecuniary damages from the co-morbidities of severe ROP and sepsis this net loss appears negligible. Conclusion: This study found that an EHD significantly decreased the odds of severe ROP and late onset sepsis; though not significant, there was a positive trend in decreasing cases of medical NEC; our surgical NEC rates dropped to 0. The benefits of human milk are vital, and the costs are nominal.
Collapse
Affiliation(s)
- Jennifer Hanford
- Division of Neonatology, Department of Child Health, Women's and Children's Hospital, University of Missouri Health Care, Columbia, Missouri, USA
| | - Kimberly Mannebach
- Division of Neonatology, Department of Child Health, Women's and Children's Hospital, University of Missouri Health Care, Columbia, Missouri, USA
| | - Adrienne Ohler
- Division of Neonatology, Department of Child Health, Women's and Children's Hospital, University of Missouri Health Care, Columbia, Missouri, USA
| | - Michael Patten
- Division of Neonatology, Department of Child Health, Women's and Children's Hospital, University of Missouri Health Care, Columbia, Missouri, USA
| | - John Pardalos
- Division of Neonatology, Department of Child Health, Women's and Children's Hospital, University of Missouri Health Care, Columbia, Missouri, USA
| |
Collapse
|
15
|
Ji YC, Sun Q, Fu CY, She X, Liu XC, He Y, Ai Q, Li LQ, Wang ZL. Exogenous Autoinducer-2 Rescues Intestinal Dysbiosis and Intestinal Inflammation in a Neonatal Mouse Necrotizing Enterocolitis Model. Front Cell Infect Microbiol 2021; 11:694395. [PMID: 34422680 PMCID: PMC8375469 DOI: 10.3389/fcimb.2021.694395] [Citation(s) in RCA: 20] [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: 04/13/2021] [Accepted: 07/21/2021] [Indexed: 12/26/2022] Open
Abstract
Autoinducer-2 (AI-2) is believed to be a bacterial interspecies signaling molecule that plays an important role in the regulation of the physiological behaviors of bacteria. The effect of AI-2 on the process of necrotizing enterocolitis (NEC) is unknown, and the aim of this study was to study the effect of AI-2 in a mouse NEC model. C57BL/6 mouse pups were randomly divided into three groups: the control group, the NEC group, and the NEC+AI-2 (NA) group. Exogenous AI-2 (500 nM) was added to the formula milk of the NA group. The concentrations of fecal AI-2 and flora were tested. The expression of cytokines, TLR4 and NF-κB in intestinal tissue was detected. The AI-2 level was significantly decreased in the NEC group (P<0.05). Compared with the NEC group, the intestinal injury scores, expression of TLR4, NF-kB, and proinflammatory factors (IL-1β, IL-6, IL-8 and TNF-α) were reduced, and expression of anti-inflammatory factor (IL-10) was increased in the NA group mice (P<0.05). At the phylum level, the Proteobacteria abundance in the NA group was significantly increased, while the Bacteroidota abundance in the control group was significantly increased (P<0.05). At the genus level, Helicobacter and Clostridium_sensu_stricto_1 exhibited significantly greater abundance in the NEC group than in the other two groups, while Lactobacillus had the opposite trend (P<0.05). In addition, the abundances of Klebsiella, Rodentibacter and Enterococcus were significantly higher in the NA group than in the NEC and control groups (P < 0.05). Exogenous AI-2 partially reverses flora disorder and decreases inflammation in an NEC mouse model.
Collapse
Affiliation(s)
- Yan-Chun Ji
- Neonatal Diagnosis and Treatment Center of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Qian Sun
- Neonatal Diagnosis and Treatment Center of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Chun-Yan Fu
- Neonatal Diagnosis and Treatment Center of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xiang She
- Neonatal Diagnosis and Treatment Center of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xiao-Chen Liu
- Neonatal Diagnosis and Treatment Center of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yu He
- Neonatal Diagnosis and Treatment Center of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Qing Ai
- Neonatal Diagnosis and Treatment Center of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Lu-Quan Li
- Neonatal Diagnosis and Treatment Center of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Zheng-Li Wang
- Neonatal Diagnosis and Treatment Center of Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
16
|
Maternal Distress and Social Support Are Linked to Human Milk Immune Properties. Nutrients 2021; 13:nu13061857. [PMID: 34072410 PMCID: PMC8226629 DOI: 10.3390/nu13061857] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 12/31/2022] Open
Abstract
Possible alterations of maternal immune function due to psychological stress may reflect immunoactive factor levels in breast milk. This study aimed to assess the association between maternal distress and breast milk levels of secretory IgA (SIgA), IgM, IgG, and lactoferrin (LF). We hypothesized that this association is moderated by maternal social support achieved from others during lactation. The study group included 103 lactating mothers and their healthy five-month-old infants. Maternal distress was determined based on the State Anxiety Inventory and the level of salivary cortisol. Social support was assessed using the Berlin Social Support Scales. Breast milk samples were collected to test for SIgA, IgM, IgG, and LF using the ELISA method. Milk immunoactive factors were regressed against maternal anxiety, social support, salivary cortisol, and infant gestational age using the general regression model. Maternal anxiety was negatively associated with milk levels of LF (β = -0.23, p = 0.028) and SIgA (β = -0.30, p = 0.004), while social support was positively associated with milk IgG (β = 0.25, p = 0.017). Neither anxiety nor social support were related to milk IgM. No association was found between the level of maternal salivary cortisol and immunoactive factors in milk. Our results suggest that maternal psychological wellbeing and social support may affect milk immune properties.
Collapse
|
17
|
Gutierrez MW, Arrieta MC. The intestinal mycobiome as a determinant of host immune and metabolic health. Curr Opin Microbiol 2021; 62:8-13. [PMID: 33993019 DOI: 10.1016/j.mib.2021.04.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 12/27/2022]
Abstract
The inclusion of fungi in recent human and animal microbiome studies has revealed that microbiome features associated with health or disease are not exclusively bacterial. Factors known to impact bacterial microbiome development, such as gestational age at birth, breast feeding status and antibiotics also impact the mycobiome. Strong inter-kingdom interactions take place in the luminal gut, and while the mycobiome exhibits increased inter-individual variability, certain fungi are stable colonizers. Here, we review recent studies showing that the gut mycobiome also plays an important role in disease states related to host immunity and energy metabolism. Some persistent species, such as Candida sp., as well as other less stable colonizers have been shown to play an important role in host-mycobiome immune cross talk. Mechanisms by which gut fungi interact with immune development have begun to be elucidated yet the majority remain elusive. Further investigation into these immune and metabolic mechanisms hold great potential for novel discoveries and will provided a much needed multi-kingdom understanding of the microbiome's influence on host health.
Collapse
Affiliation(s)
- Mackenzie W Gutierrez
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada; Department of Pediatrics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; International Microbiome Center, University of Calgary, AB, Canada
| | - Marie-Claire Arrieta
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada; Department of Pediatrics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; International Microbiome Center, University of Calgary, AB, Canada.
| |
Collapse
|
18
|
Morais J, Marques C, Faria A, Teixeira D, Barreiros-Mota I, Durão C, Araújo J, Ismael S, Brito S, Cardoso M, Macedo I, Pereira E, Tomé T, Calhau C. Influence of Human Milk on Very Preterms' Gut Microbiota and Alkaline Phosphatase Activity. Nutrients 2021; 13:1564. [PMID: 34066473 PMCID: PMC8148101 DOI: 10.3390/nu13051564] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 01/12/2023] Open
Abstract
The FEEDMI Study (NCT03663556) evaluated the influence of infant feeding (mother's own milk (MOM), donor human milk (DHM) and formula) on the fecal microbiota composition and alkaline phosphatase (ALP) activity in extremely and very preterm infants (≤32 gestational weeks). In this observational study, preterm infants were recruited within the first 24 h after birth. Meconium and fecal samples were collected at four time points (between the 2nd and the 26th postnatal days. Fecal microbiota was analyzed by RT-PCR and by 16S rRNA sequencing. Fecal ALP activity, a proposed specific biomarker of necrotizing enterocolitis (NEC), was evaluated by spectrophotometry at the 26th postnatal day. A total of 389 fecal samples were analyzed from 117 very preterm neonates. Human milk was positively associated with beneficial bacteria, such as Bifidobacterium, Bacteroides ovatus, and Akkermancia muciniphila, as well as bacterial richness. Neonates fed with human milk during the first week of life had increased Bifidobacterium content and fecal ALP activity on the 26th postnatal day. These findings point out the importance of MOM and DHM in the establishment of fecal microbiota on neonates prematurely delivered. Moreover, these results suggest an ALP pathway by which human milk may protect against NEC.
Collapse
Affiliation(s)
- Juliana Morais
- Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (J.M.); (C.M.); (A.F.); (D.T.); (I.B.-M.); (C.D.); (J.A.); (S.I.)
- CHRC-Comprehensive Health Research Centre, CEDOC-Chronic Diseases Research Center, Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Cláudia Marques
- Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (J.M.); (C.M.); (A.F.); (D.T.); (I.B.-M.); (C.D.); (J.A.); (S.I.)
- CINTESIS-Center for Health Technology Services Research, Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Ana Faria
- Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (J.M.); (C.M.); (A.F.); (D.T.); (I.B.-M.); (C.D.); (J.A.); (S.I.)
- CHRC-Comprehensive Health Research Centre, CEDOC-Chronic Diseases Research Center, Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Diana Teixeira
- Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (J.M.); (C.M.); (A.F.); (D.T.); (I.B.-M.); (C.D.); (J.A.); (S.I.)
- CHRC-Comprehensive Health Research Centre, CEDOC-Chronic Diseases Research Center, Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
- NOVA Medical School, Unidade Universitária Lifestyle Medicine José de Mello Saúde, 1169-056 Lisboa, Portugal
| | - Inês Barreiros-Mota
- Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (J.M.); (C.M.); (A.F.); (D.T.); (I.B.-M.); (C.D.); (J.A.); (S.I.)
- CHRC-Comprehensive Health Research Centre, CEDOC-Chronic Diseases Research Center, Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Catarina Durão
- Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (J.M.); (C.M.); (A.F.); (D.T.); (I.B.-M.); (C.D.); (J.A.); (S.I.)
- EPIUnit-Institute of Public Health, Universidade do Porto, 4050-600 Porto, Portugal
| | - João Araújo
- Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (J.M.); (C.M.); (A.F.); (D.T.); (I.B.-M.); (C.D.); (J.A.); (S.I.)
- CINTESIS-Center for Health Technology Services Research, Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Shámila Ismael
- Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (J.M.); (C.M.); (A.F.); (D.T.); (I.B.-M.); (C.D.); (J.A.); (S.I.)
- CHRC-Comprehensive Health Research Centre, CEDOC-Chronic Diseases Research Center, Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Sara Brito
- Pediatrics Department, Maternidade Dr. Alfredo da Costa, Centro Hospitalar Universitário de Lisboa Central, 2890-495 Lisboa, Portugal; (S.B.); (I.M.); (E.P.); (T.T.)
| | - Manuela Cardoso
- Nutrition and Dietetics Unit, Maternidade Dr. Alfredo da Costa, Centro Hospitalar Universitário de Lisboa Central, 2890-495 Lisboa, Portugal;
| | - Israel Macedo
- Pediatrics Department, Maternidade Dr. Alfredo da Costa, Centro Hospitalar Universitário de Lisboa Central, 2890-495 Lisboa, Portugal; (S.B.); (I.M.); (E.P.); (T.T.)
| | - Esmeralda Pereira
- Pediatrics Department, Maternidade Dr. Alfredo da Costa, Centro Hospitalar Universitário de Lisboa Central, 2890-495 Lisboa, Portugal; (S.B.); (I.M.); (E.P.); (T.T.)
| | - Teresa Tomé
- Pediatrics Department, Maternidade Dr. Alfredo da Costa, Centro Hospitalar Universitário de Lisboa Central, 2890-495 Lisboa, Portugal; (S.B.); (I.M.); (E.P.); (T.T.)
| | - Conceição Calhau
- Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (J.M.); (C.M.); (A.F.); (D.T.); (I.B.-M.); (C.D.); (J.A.); (S.I.)
- CINTESIS-Center for Health Technology Services Research, Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
- NOVA Medical School, Unidade Universitária Lifestyle Medicine José de Mello Saúde, 1169-056 Lisboa, Portugal
| |
Collapse
|
19
|
Jiang YN, Ye YX, Sangild PT, Thymann T, Engelsen SB, Khakimov B, Jiang PP. Plasma Metabolomics to Evaluate Progression of Necrotising Enterocolitis in Preterm Pigs. Metabolites 2021; 11:metabo11050283. [PMID: 33946896 PMCID: PMC8146597 DOI: 10.3390/metabo11050283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/19/2021] [Accepted: 04/26/2021] [Indexed: 02/04/2023] Open
Abstract
Necrotising enterocolitis (NEC) is a microbiome-dependent gut disease in preterm infants in early life. Antibiotic treatment is a common intervention for NEC. How NEC lesions, with or without antibiotics, affect plasma metabolome was explored in this study. Formula-fed preterm pigs were used as a model for human NEC and treated with saline, parenteral or oral antibiotics (n = 15-17) for four days after delivery. Gut tissues were collected for evaluation of NEC-like lesions and plasma for metabolomic analysis by proton nuclear magnetic resonance spectroscopy (1H-NMR). Metabolites were annotated, quantified and subjected to statistical modelling to delineate the effects of NEC and antibiotic treatment. Presence of severe NEC lesions, not antibiotic treatment, was the main drive for plasma metabolite changes. Relative to other pigs, pigs with severe NEC lesions had higher levels of alanine, histidine and myo-inositol, and lower levels of 3-hydroxybutyric acid and isobutyric acid. Across NEC lesion states (healthy, mild, severe), antibiotics directly affected only a few metabolites (tryptophan, 3-phenyllactic acid). Together and independently, NEC and antibiotic treatment affected circulating metabolites in preterm pigs. Amino acids and plasma metabolites, partly related to the gut microbiome, may be helpful to monitor progression of NEC lesions after proper validation.
Collapse
Affiliation(s)
- Yan-Nan Jiang
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Y.-N.J.); (Y.-X.Y.)
| | - Yong-Xin Ye
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Y.-N.J.); (Y.-X.Y.)
| | - Per Torp Sangild
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark; (P.T.S.); (T.T.)
- Department of Neonatology, Rigshospitalet, DK-2100 Copenhagen, Denmark
- Department of Paediatrics, Odense University Hospital, DK-5000 Odense, Denmark
| | - Thomas Thymann
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark; (P.T.S.); (T.T.)
| | - Søren Balling Engelsen
- Department of Food Science, University of Copenhagen, DK-1958 Frederiksberg, Denmark; (S.B.E.); (B.K.)
| | - Bekzod Khakimov
- Department of Food Science, University of Copenhagen, DK-1958 Frederiksberg, Denmark; (S.B.E.); (B.K.)
| | - Ping-Ping Jiang
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Y.-N.J.); (Y.-X.Y.)
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark; (P.T.S.); (T.T.)
- Correspondence:
| |
Collapse
|
20
|
Chen W, Yang C, Xue H, Huang Q. The protective effect and mechanism of epidermal growth factor on necrotizing enterocolitis in a neonatal rat model. Transl Pediatr 2021; 10:900-913. [PMID: 34012839 PMCID: PMC8107864 DOI: 10.21037/tp-21-81] [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: 01/28/2021] [Accepted: 04/21/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is the most common acquired gastrointestinal emergency in premature infants. This study aimed to investigate the protective effect and mechanism of epidermal growth factor (EGF) on NEC in a neonatal rat model. METHODS We randomly divided 50 newborn SD rats into a control group, NEC group, NEC + 50 ng/mL EGF group, NEC + 500 ng/mL EGF group, and NEC + 1,000 ng/mL EGF group, with 10 cases in each group. The appearance of intestinal tissue, physiological status score, inflammatory factor level, HE staining, and pathological score were used to evaluate the protective effect. A one cm tissue sample from the proximal ileum of the ileocecal area of five rats from the NEC group and the group that showed a significant protective effect were extracted for transcriptome sequencing. RESULTS The levels of IL-1β and IL-6 in the intestinal mucosa in the NEC + 500 ng/mL EGF group were significantly lower than those in the NEC + 1,000 ng/mL EGF group (P<0.05). Transcriptome sequencing suggested that EGF effects the intestinal barrier, apoptosis, and inflammation of the NEC intestine. CONCLUSIONS We conclude that the oral administration of 500 ng/mL EGF effectively inhibits intestinal inflammation in NEC neonatal rat models, thereby affecting the barrier function of the intestinal tract.
Collapse
Affiliation(s)
- Wenqian Chen
- Department of Neonatology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Changyi Yang
- Department of Neonatology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Heng Xue
- Department of Laboratory Medicine, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Qi Huang
- Department of Neonatology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
21
|
Chen Y, Wang Z, Xiao D, Zeng H, Ma X. Predicting the Severity of Acute Appendicitis of Young Children (<3 Years Old): Development and Assessment of a New Prediction Nomogram. Front Pediatr 2021; 9:763125. [PMID: 34869120 PMCID: PMC8637160 DOI: 10.3389/fped.2021.763125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/11/2021] [Indexed: 12/29/2022] Open
Abstract
Objective: There is a lack of assessment methods of acute appendicitis in little children. The purpose of this study was to develop and internally validate a nomogram for predicting the severity of acute appendicitis of young children (<3 years old). Methods: We develop a prediction model based on a training dataset of 121 patients (<3 years old) with acute appendicitis. Admission information was collected between January 2010 and January 2021, which contained demographic characteristic, laboratory examinations, treatment and pathology type, etc. Logistic regression analysis was used to identify independent risk factors and establish the predictive model. C-index and calibration curves were applied to evaluate the performance of the nomogram. Then corrected C-index was calculated to conduct internal verification by using the bootstrapping validation. Decision curve analysis determined clinical application of the prediction model. Results: Predictors contained in the prediction nomogram included weight for age, onset time (from developing symptoms to hospital), admission temperature, leukocyte count, neutrophil ratio, and total bilirubin. Logistic regression analysis showed that weight for age (X1) < -2.32 SD (P = 0.046), onset time (X2) > 2.5 days (P = 0.044), admission temperature (X3) > 38.5°C (P = 0.009), leukocyte count (X4) > 12.185*109/L (P = 0.045), neutrophil ratio (X5) > 68.7% (P = 0.029), and total bilirubin (X6) > 9.05 μmol/L (P = 0.035) were found to be significant for predicting the severity of appendicitis. The logistic regression equation was logit (P) = -0.149X1 + 0.51X2 + 1.734X3 + 0.238X4 + 0.061X5 + 0.098X6 - 75.229. C-index of nomogram was calculated at 0.8948 (95% Cl: 0.8332-0.9567) and it still was 0.8867 through bootstrapping validation. Decision curve analysis showed that when the threshold probability ranged from 14 to 88%, there is a net benefit of using this prediction model for severity of appendicitis in little children. Conclusion: This novel nomogram incorporating the weight for age, onset time, admission temperature, leukocyte count, neutrophil ratio, and total bilirubin could be conveniently used to estimate the severity of appendicitis of young children <3 years old) and determine appropriate treatment options in time.
Collapse
Affiliation(s)
- Yang Chen
- Shenzhen Children's Hospital, Shenzhen, China.,College of Medicine, Shantou University, Shantou, China
| | | | - Dong Xiao
- Shenzhen Children's Hospital, Shenzhen, China
| | - Hongwu Zeng
- Shenzhen Children's Hospital, Shenzhen, China
| | - Xiaopeng Ma
- Shenzhen Children's Hospital, Shenzhen, China
| |
Collapse
|
22
|
Stevenson DK, Wong RJ, Hay WW. Comments on the 20 th Anniversary of NeoReviews. Neoreviews 2020; 21:e643-e648. [PMID: 33004557 DOI: 10.1542/neo.21-10-e643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Affiliation(s)
- David K Stevenson
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA
| | - Ronald J Wong
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA
| | | |
Collapse
|
23
|
Czosnykowska-Łukacka M, Lis-Kuberka J, Królak-Olejnik B, Orczyk-Pawiłowicz M. Changes in Human Milk Immunoglobulin Profile During Prolonged Lactation. Front Pediatr 2020; 8:428. [PMID: 32850542 PMCID: PMC7426452 DOI: 10.3389/fped.2020.00428] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 06/19/2020] [Indexed: 01/09/2023] Open
Abstract
Mother's milk immunoglobulins (Igs) delivered to infants during breastfeeding are crucial in shaping and modulating immature infants' immune system and provide efficient protection against pathogens. The aim of the study was to evaluate the immunoglobulin concentrations in milk of 116 lactating mothers over prolonged lactation from the 1st to the 48th month using the ELISA method. The concentration of proteins, SIgA and IgG, but not IgM, showed a positive correlation (r = 0.69, p < 0.005; r = 0.54, p < 0.05; and r = 0.27, p < 0.05, respectively) with lactation from the 1st to the 48th month. The lowest concentrations of SIgA and IgG were observed for the first year (2.12 ± 0.62 g/L and 14.71 ± 6.18 mg/L, respectively) and the highest after the 2nd year of lactation (7.55 ± 7.16 g/L and 18.95 ± 6.76 mg/L, respectively). The IgM concentration remained stable during 2 years (2.81 ± 2.74 mg/L), but after 24 months it was higher (3.82 ± 3.05 mg/L), although not significantly. Moreover, negative correlations of protein (r = -0.24, p < 0.05) and SIgA (r = -0.47, p < 0.05) concentrations with the number of feedings were found. Human milk after the 2nd year of lactation contains significantly higher concentrations of protein, SIgA, and IgG. High concentration of immunoglobulins and protein during prolonged lactation is an additional argument to support breastfeeding even after introducing solid foods and should be one of the overarching goals in the protection of children's health.
Collapse
Affiliation(s)
| | - Jolanta Lis-Kuberka
- Department of Chemistry and Immunochemistry, Wroclaw Medical University, Wrocław, Poland
| | | | | |
Collapse
|
24
|
Sampah MES, Hackam DJ. Dysregulated Mucosal Immunity and Associated Pathogeneses in Preterm Neonates. Front Immunol 2020; 11:899. [PMID: 32499778 PMCID: PMC7243348 DOI: 10.3389/fimmu.2020.00899] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 04/17/2020] [Indexed: 12/12/2022] Open
Abstract
Many functions of the immune system are impaired in neonates, allowing vulnerability to serious bacterial, viral and fungal infections which would otherwise not be pathogenic to mature individuals. This vulnerability is exacerbated in compromised newborns such as premature neonates and those who have undergone surgery or who require care in an intensive care unit. Higher susceptibility of preterm neonates to infections is associated with delayed immune system maturation, with deficiencies present in both the innate and adaptive immune components. Here, we review recent insights into early life immunity, and highlight features associated with compromised newborns, given the challenges of studying neonatal immunity in compromised neonates due to the transient nature of this period of life, and logistical and ethical obstacles posed by undertaking studies newborns and infants. Finally, we highlight how the unique immunological characteristics of the premature host play key roles in the pathogenesis of diseases that are unique to this population, including necrotizing enterocolitis and the associated sequalae of lung and brain injury.
Collapse
Affiliation(s)
- Maame Efua S Sampah
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - David J Hackam
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
25
|
Carbone F, Montecucco F, Sahebkar A. Current and emerging treatments for neonatal sepsis. Expert Opin Pharmacother 2020; 21:549-556. [PMID: 32011188 DOI: 10.1080/14656566.2020.1721464] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 01/22/2020] [Indexed: 01/27/2023]
Abstract
Introduction: Mortality due to sepsis is still prevalent, peaking at extreme ages of life including infancy. Despite many efforts, the peculiarity of the infant immune system has limited further advances in its treatment. Indeed, neonates experience a dramatic physiological transition from immune tolerance to the maternal antigens to functional maturity. Such a transition is extremely dynamic, as is the pathophysiology of infant sepsis, which is dependent on many infant, maternal, and environmental factors.Areas covered: In this review, the authors critically update and summarize the current paradigm of immunomodulation in infant sepsis. They confirm how exogenous stimulation of the immune system through intravenous immunoglobulin, colony stimulating factors, and granulocyte transfusion have failed to impact on the prognosis of infant sepsis. They also strongly support the beneficial effects of supplementation/replacement therapies with products naturally contained within maternal milk as well as antioxidant compounds.Expert opinion: Breastfeeding is beneficial against sepsis. Knowledge of the neonatal immune system is indeed too limited to effectively strengthen immune response by exogenous interventions, especially in preterm and low-birth-weight infants. Awareness of this limitation should pave the way for future studies (e.g. gender- and omics-based) aimed at better characterizing the infant immune system and promoting a more tailored approach.
Collapse
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
26
|
Neonatal Microbiome and Its Relationship to Necrotizing Enterocolitis: A Review of the Science. J Perinat Neonatal Nurs 2020; 34:276-282. [PMID: 32697548 DOI: 10.1097/jpn.0000000000000507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Necrotizing enterocolitis (NEC) occurs in many premature infants hospitalized in the neonatal intensive care unit. About 3% to 15% of very low-weight premature infants develop NEC, with an estimated 30% mortality rate for the cases requiring surgery. Currently, there is no known pathogenesis for NEC in the patient's populations. However, one of the most widely accepted hypotheses is having an abnormal fetal gut microbiome. The purpose of this review is to discuss some current methods of dysbiosis in the neonatal microbiome, such as maternal health, breastfeeding, and delivery method, and then to connect these to the occurrence of NEC in the infant and finally discuss some possibilities for limiting the occurrence of NEC in the future.
Collapse
|
27
|
Abstract
Progress in our understanding of the pathophysiology, prevention and treatment of necrotizing enterocolitis (NEC) has been hampered for many reasons. Included among these is the fact that what we are calling "NEC" is likely to represent different disease processes, which need to be delineated before evaluating individual pathogenic mechanisms and attempting to develop predictive and diagnostic biomarkers. Treatment is also likely to be hampered because not all of the different entities called "NEC" will respond to the same regimen. In this review, some of these entities will be discussed in more detail, with suggestions for refining our approach toward improving methods for their diagnosis, prevention and treatment.
Collapse
Affiliation(s)
- Josef Neu
- Pediatrics/Neonatology, University of Florida, Gainesville, Florida, USA,
| |
Collapse
|