1
|
Núñez-Ramos R, Escuder-Vieco D, Rico Cruz C, Diego-Poncela CD, Vázquez-Román S, Germán-Díaz M, García-Lara NR, Pallás-Alonso C. Bioelectrical Impedance Vector Analysis in Extremely Low-Birth-Weight Infants to Assess Nutritional Status: Breakthroughs and Insights. Nutrients 2024; 16:4348. [PMID: 39770969 PMCID: PMC11677361 DOI: 10.3390/nu16244348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: To obtain bioelectrical data to assess nutritional status for extremely low-birth-weight (ELBW) infants upon reaching term-corrected age. Methods: A descriptive, observational, prospective, and single-center study, which included ELBW preterm infants was performed. The study variables collected were gestational age, sex, and anthropometry at birth and at term-corrected age. Bioelectrical impedance vector analysis (BIVA) was performed by a phase-sensitive device (BIA 101 BIVA PRO AKERN srl, Pisa, Italy). The components of the impedance vector-resistance (R) and reactance (Xc)-were normalized for body height (H). For each subject, the measurement was taken between the 36th and 44th weeks of postmenstrual age (PMA). A semi-quantitative analysis of body composition was performed using the vector modality of the BIA. Using the RXc graph method, the bivariate 95% confidence intervals of the mean vectors were constructed. From the bivariate normal distribution of R/H and Xc/H, the bivariate 95%, 75%, and 50% tolerance intervals for this cohort were drawn. The individual impedance vectors were compared with the distribution of the vectors from other populations. Results: 85 ELBW infants (40 male, 45 female) were included, with a mean gestational age at birth of 26 + 6 weeks (±1.76). Mean R/H was 870.33 (±143.21) Ohm/m and Xc/H was 86.84 (±19.05) Ohm/m. We found differences in the bioelectrical data with regard to gender, with resistance values being significantly higher in females. Our ellipses align closely with those from other term neonatal cohorts. Conclusions: Bioelectrical data and the confidence and tolerance ellipses of an ELBW infant cohort are presented and can be used as a reference standard for nutritional assessment at discharge.
Collapse
Affiliation(s)
- Raquel Núñez-Ramos
- Department of Pediatrics, 12 de Octubre University Hospital, 28041 Madrid, Spain
| | - Diana Escuder-Vieco
- Aladina-MGU-Regional Human Milk Bank, 12 de Octubre University Hospital, 28041 Madrid, Spain; (D.E.-V.); (C.R.C.); (C.D.D.-P.); (S.V.-R.); (N.R.G.-L.); (C.P.-A.)
- Research Institute i+12, 12 de Octubre University Hospital, 28041 Madrid, Spain
| | - Carolina Rico Cruz
- Aladina-MGU-Regional Human Milk Bank, 12 de Octubre University Hospital, 28041 Madrid, Spain; (D.E.-V.); (C.R.C.); (C.D.D.-P.); (S.V.-R.); (N.R.G.-L.); (C.P.-A.)
- Department of Neonatology, 12 de Octubre University Hospital, 28041 Madrid, Spain
| | - Cristina De Diego-Poncela
- Aladina-MGU-Regional Human Milk Bank, 12 de Octubre University Hospital, 28041 Madrid, Spain; (D.E.-V.); (C.R.C.); (C.D.D.-P.); (S.V.-R.); (N.R.G.-L.); (C.P.-A.)
- Department of Neonatology, 12 de Octubre University Hospital, 28041 Madrid, Spain
| | - Sara Vázquez-Román
- Aladina-MGU-Regional Human Milk Bank, 12 de Octubre University Hospital, 28041 Madrid, Spain; (D.E.-V.); (C.R.C.); (C.D.D.-P.); (S.V.-R.); (N.R.G.-L.); (C.P.-A.)
- Research Institute i+12, 12 de Octubre University Hospital, 28041 Madrid, Spain
- Department of Neonatology, 12 de Octubre University Hospital, 28041 Madrid, Spain
| | - Marta Germán-Díaz
- Department of Pediatric Nutrition, 12 de Octubre University Hospital, 28041 Madrid, Spain;
| | - Nadia Raquel García-Lara
- Aladina-MGU-Regional Human Milk Bank, 12 de Octubre University Hospital, 28041 Madrid, Spain; (D.E.-V.); (C.R.C.); (C.D.D.-P.); (S.V.-R.); (N.R.G.-L.); (C.P.-A.)
- Research Institute i+12, 12 de Octubre University Hospital, 28041 Madrid, Spain
- Department of Neonatology, 12 de Octubre University Hospital, 28041 Madrid, Spain
| | - Carmen Pallás-Alonso
- Aladina-MGU-Regional Human Milk Bank, 12 de Octubre University Hospital, 28041 Madrid, Spain; (D.E.-V.); (C.R.C.); (C.D.D.-P.); (S.V.-R.); (N.R.G.-L.); (C.P.-A.)
- Research Institute i+12, 12 de Octubre University Hospital, 28041 Madrid, Spain
- Department of Neonatology, 12 de Octubre University Hospital, 28041 Madrid, Spain
| |
Collapse
|
2
|
Mishima S, Mitsui T, Tani K, Maki J, Eto E, Hayata K, Washio Y, Yoshimoto J, Tsukahara H, Masuyama H. Short stature in small-for-gestational-age offspring born to mothers with hypertensive disorders of pregnancy. Hypertens Pregnancy 2023; 42:2187623. [PMID: 36891613 DOI: 10.1080/10641955.2023.2187623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
Abstract
OBJECTIVE To investigate the incidence and risk factors of small-for-gestational age (SGA) short stature at 2 and 3 years of age in SGA offspring born to women with hypertensive disorders of pregnancy (HDP). METHODS We examined 226 women with HDP whose respective SGA offspring were delivered. RESULTS Eighty offspring (41.2%) were diagnosed with SGA short stature. The prematurity before 32 weeks of gestation was the most significant factor for catch-up growth failure. CONCLUSION In SGA offspring born to women with HDP, SGA short stature incidence was high, and the risk factor was prematurity before 32 weeks of gestation.
Collapse
Affiliation(s)
- Sakurako Mishima
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Takashi Mitsui
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Kazumasa Tani
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Jota Maki
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Eriko Eto
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Kei Hayata
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Yosuke Washio
- Department of Pediatrics, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Junko Yoshimoto
- Department of Pediatrics, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hirokazu Tsukahara
- Department of Pediatrics, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hisashi Masuyama
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Okayama, Japan
| |
Collapse
|
3
|
Strobel KM, Del Vecchio G, Devaskar SU, Calkins KL. Gut Microbes and Circulating Cytokines in Preterm Infants with Growth Failure. J Nutr 2023; 153:120-130. [PMID: 36913445 PMCID: PMC10196572 DOI: 10.1016/j.tjnut.2022.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 10/12/2022] [Accepted: 10/23/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Growth failure (GF) is a multifactorial problem in preterm infants. The intestinal microbiome and inflammation may contribute to GF. OBJECTIVES This study's objective was to compare the gut microbiome and plasma cytokines in preterm infants with and without GF. METHODS This was a prospective cohort study of infants with birth weights of <1750 g. Infants with a weight or length z-score change from birth to discharge or death that was less than or equal to -0.8 (GF group) were compared with infants without GF [control (CON) group]. The primary outcome was the gut microbiome (at weeks 1-4 of age), assessed by 16S rRNA gene sequencing using Deseq2. Secondary outcomes included inferred metagenomic function and plasma cytokines. Phylogenetic Investigation of Communities by Reconstruction of Unobserved States determined metagenomic function, which was compared using ANOVA. Cytokines were measured by 2-multiplexed immunometric assays and compared using Wilcoxon tests and linear mixed models. RESULTS GF (n = 14) and CON group (n = 13) had similar median (IQR) birth weight (1380 [780-1578] g vs. 1275 [1013-1580] g) and gestational age (29 [25-31] weeks vs. 30 [29-32] weeks). Compared with the CON group, the GF group had a greater abundance of Escherichia/Shigella in weeks 2 and 3, Staphylococcus in week 4, and Veillonella in weeks 3 and 4 (P-adjusted < 0.001 for all). Plasma cytokine concentrations did not differ significantly between the cohorts. When all time points are combined, fewer microbes were involved in TCA cycle activity in the GF group compared with the CON group (P = 0.023). CONCLUSIONS In this study, when compared with CON infants, GF infants had a distinct microbial signature with increased Escherichia/Shigella and Firmicutes and fewer microbes associated with energy production at later weeks of hospitalization. These findings may suggest a mechanism for aberrant growth.
Collapse
Affiliation(s)
- Katie M Strobel
- Department of Pediatrics, Division of Neonatology and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA; The University of California Los Angeles Children's Discovery and Innovation Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Giorgia Del Vecchio
- Department of Pediatrics, Division of Neonatology and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA; The University of California Los Angeles Children's Discovery and Innovation Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Sherin U Devaskar
- Department of Pediatrics, Division of Neonatology and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA; The University of California Los Angeles Children's Discovery and Innovation Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Kara L Calkins
- Department of Pediatrics, Division of Neonatology and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA; The University of California Los Angeles Children's Discovery and Innovation Institute, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
4
|
Khan A, Mi H, Gao F, Hu Q, Gu X, Ma F, Qu L, Li S, Dai Y, Hao H. Dynamic changes of the gut microbial colonization in preterm infants with different time points after birth. Front Microbiol 2023; 14:1078426. [PMID: 36876108 PMCID: PMC9983350 DOI: 10.3389/fmicb.2023.1078426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/12/2023] [Indexed: 02/19/2023] Open
Abstract
Risks associated with preterm birth are unevenly distributed across all gestations. At earlier gestational ages, complications such as necrotizing enterocolitis (NEC) and late-onset sepsis (LOS) conditions are significantly more common and are associated with a shift in the composition of the gut microbiome. Conventional bacterial culture techniques demonstrate that the colonization of the gut microbiota of preterm infants differs significantly from that of healthy-term infants. The current study aimed to investigate the impact of preterm infancy on the dynamic changes of fecal microbiota in preterm infants at different time points (1, 7, 14, 21, 28, and 42 days) after birth. We selected 12 preterm infants hospitalized in the Sixth Affiliated Hospital of Sun Yat-sen University from January 2017 to December 2017. A total of 130 fecal specimens from preterm infants were analyzed using 16S rRNA gene sequencing. We found that the colonization process of fecal microbiota in preterm infants is highly dynamic at different time points after birth, i.e., Exiguobacterium, Acinetobacter, and Citrobacter showed a declining abundance pattern with the advancement of age, while the bacterial groups of Enterococcus (Klebsiella and Escherichia coli) gradually grew and became the main microbiota during the development of fecal microbiota in preterm infants at the age of 42 days. Furthermore, the colonization of intestinal Bifidobacteria in preterm infants was relatively late and did not rapidly become the predominant microbiota. Moreover, the results also showed the presence of Chryseobacterium bacterial group, whose colonization was different in different time point groups. Conclusively, our findings deepen our comprehension and offer new perspectives on targeting particular bacteria in the treatment of preterm infants at different time points after birth.
Collapse
Affiliation(s)
- Adnan Khan
- Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Hongying Mi
- Department of Pediatric, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Fei Gao
- Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.,Section of Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Qi Hu
- NEOMICS Institute, Shenzhen, China
| | - Xia Gu
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Fei Ma
- Department of Pediatrics, Zhuhai Women and Children's Hospital, Zhuhai, China
| | - LiuHong Qu
- Department of Neonatology, The Maternal and Child Health Care Hospital of HuaDu District, Guangzhou, China
| | - Sitao Li
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yiheng Dai
- Department of Neonatology, Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Hu Hao
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Xu J, Shin J, McGee M, Unger S, Bando N, Sato J, Vandewouw M, Patel Y, Branson HM, Paus T, Pausova Z, O'Connor DL. Intake of mother's milk by very-low-birth-weight infants and variation in DNA methylation of genes involved in neurodevelopment at 5.5 years of age. Am J Clin Nutr 2022; 116:1038-1048. [PMID: 35977396 PMCID: PMC9535521 DOI: 10.1093/ajcn/nqac221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 08/09/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Mechanisms responsible for associations between intake of mother's milk in very-low-birth-weight (VLBW, <1500 g) infants and later neurodevelopment are poorly understood. It is proposed that early nutrition may affect neurodevelopmental pathways by altering gene expression through epigenetic modification. Variation in DNA methylation (DNAm) at cytosine-guanine dinucleotides (CpGs) is a commonly studied epigenetic modification. OBJECTIVES We aimed to assess whether early mother's milk intake by VLBW infants is associated with variations in DNAm at 5.5 y, and whether these variations correlate with neurodevelopmental phenotypes. METHODS This cohort study was a 5.5-y follow-up (2016-2018) of VLBW infants born in Ontario, Canada who participated in the Donor Milk for Improved Neurodevelopmental Outcomes trial. We performed an epigenome-wide association study (EWAS) to test whether percentage mother's milk (not including supplemental donor milk) during hospitalization was associated with DNAm in buccal cells during early childhood (n = 143; mean ± SD age: 5.7 ± 0.2 y; birth weight: 1008 ± 517 g). DNAm was assessed with the Illumina Infinium MethylationEPIC array at 814,583 CpGs. In secondary analyses, we tested associations between top-ranked CpGs and measures of early childhood neurodevelopment, e.g., total surface area of the cerebral cortex (n = 41, MRI) and Full-Scale IQ (n = 133, Wechsler Preschool and Primary Scale of Intelligence-IV). RESULTS EWAS analysis demonstrated percentage mother's milk intake by VLBW infants during hospitalization was associated with DNAm at 2 CpGs, cg03744440 [myosin XVB (MYO15B)] and cg00851389 [metallothionein 1A (MT1A)], at 5.5 y (P < 9E-08). Gene set enrichment analysis indicated that top-ranked CpGs (P < 0.001) were annotated to genes enriched in neurodevelopmental biological processes. Corroborating these findings, DNAm at several top identified CpGs from the EWAS was associated with cortical surface area and IQ at 5.5 y (P < 0.05). CONCLUSIONS In-hospital percentage mother's milk intake by VLBW infants was associated with variations in DNAm of neurodevelopmental genes at 5.5 y; some of these DNAm variations are associated with brain structure and IQ.This trial was registered at isrctn.com as ISRCTN35317141 and at clinicaltrials.gov as NCT02759809.
Collapse
Affiliation(s)
- Jingxiong Xu
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Jean Shin
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Meghan McGee
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Sharon Unger
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
- Department of Pediatrics, Sinai Health, Toronto, Ontario, Canada
- Division of Neonatology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nicole Bando
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Julie Sato
- Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Neuroscience & Mental Health Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Marlee Vandewouw
- Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada
- Neuroscience & Mental Health Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Autism Research Centre, Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Yash Patel
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Helen M Branson
- Division of Neuroradiology, Department of Medical Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada
| | - Tomas Paus
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Department of Psychiatry, Faculty of Medicine and CHU Sainte-Justine, University of Montreal, Montreal, Quebec, Canada
- Department of Neuroscience, Faculty of Medicine and CHU Sainte-Justine, University of Montreal, Montreal, Quebec, Canada
| | - Zdenka Pausova
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Deborah L O'Connor
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Pediatrics, Sinai Health, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Inada A, Inada O. Differences in long-term effects of standard rodent diets on blood glucose and body weight of offspring. Diabetol Int 2022; 13:615-623. [PMID: 36117929 PMCID: PMC9477981 DOI: 10.1007/s13340-022-00578-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/10/2022] [Indexed: 01/03/2023]
Abstract
Standard rodent diets are similar and contain well-balanced components, such as crude protein, fat, fiber, and ash. However, it is not clear whether there are differences in their long-term effects on metabolism. Here, we investigated the effects of long-term feeding of major standard diets, CRF-1, CE-2, and FR-1 to wild-type (WT) C57BL/6 mice on the blood glucose levels and body weight gain of their offspring, which were raised on the same diet and in the same environment as the mothers. The offspring have been influenced by the maternal diet during the fetal and neonatal stages, and were maintained on the same diet thereafter (until 60 weeks of age). In the CE-2 group, the offspring showed stable blood glucose levels and had the lowest body weight. The FR1 group showed the lowest blood glucose level, but body weight was increased significantly compared to the CE-2 group. In the CRF-1 group, higher blood glucose levels were seen from the neonatal stage and body weight increased more rapidly than in the other groups. Next, to determine the effects of blood glucose level and housing pattern on food and water consumption, severely diabetic (hyperglycemic) inducible cAMP early repressor (ICER) transgenic (Tg) mice and littermate WT mice (normoglycemic) were fed CE-2 diet and housed individually or in groups. Food and water consumption of WT mice was independent of housing pattern, whereas Tg mice showed significantly increased food and water consumption when housed individually, compared to group housing, and this did not change at different ages. Thus, the selection of standard diet and rearing method can have a marked impact on experimental outcomes in experiments using mice and presumably mouse cells, especially in studies of metabolism, diabetes, and obesity.
Collapse
Affiliation(s)
- Akari Inada
- grid.177174.30000 0001 2242 4849Diabetes and Genes, Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- grid.417982.10000 0004 0623 246XPresent Address: Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation (IBRI), 6-3-7 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047 Japan
| | - Oogi Inada
- grid.177174.30000 0001 2242 4849Diabetes and Genes, Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
7
|
Melnik BC, Schmitz G. Milk Exosomal microRNAs: Postnatal Promoters of β Cell Proliferation but Potential Inducers of β Cell De-Differentiation in Adult Life. Int J Mol Sci 2022; 23:ijms231911503. [PMID: 36232796 PMCID: PMC9569743 DOI: 10.3390/ijms231911503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic β cell expansion and functional maturation during the birth-to-weaning period is driven by epigenetic programs primarily triggered by growth factors, hormones, and nutrients provided by human milk. As shown recently, exosomes derived from various origins interact with β cells. This review elucidates the potential role of milk-derived exosomes (MEX) and their microRNAs (miRs) on pancreatic β cell programming during the postnatal period of lactation as well as during continuous cow milk exposure of adult humans to bovine MEX. Mechanistic evidence suggests that MEX miRs stimulate mTORC1/c-MYC-dependent postnatal β cell proliferation and glycolysis, but attenuate β cell differentiation, mitochondrial function, and insulin synthesis and secretion. MEX miR content is negatively affected by maternal obesity, gestational diabetes, psychological stress, caesarean delivery, and is completely absent in infant formula. Weaning-related disappearance of MEX miRs may be the critical event switching β cells from proliferation to TGF-β/AMPK-mediated cell differentiation, whereas continued exposure of adult humans to bovine MEX miRs via intake of pasteurized cow milk may reverse β cell differentiation, promoting β cell de-differentiation. Whereas MEX miR signaling supports postnatal β cell proliferation (diabetes prevention), persistent bovine MEX exposure after the lactation period may de-differentiate β cells back to the postnatal phenotype (diabetes induction).
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
- Correspondence: ; Tel.: +49-52-4198-8060
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, University of Regensburg, D-93053 Regensburg, Germany
| |
Collapse
|
8
|
Kindt A, Kraus Y, Rasp D, Foerster KM, Ahmidi N, Flemmer AW, Herber-Jonat S, Heinen F, Weigand H, Hankemeier T, Koletzko B, Krumsiek J, Babl J, Hilgendorff A. Improved Macro- and Micronutrient Supply for Favorable Growth and Metabolomic Profile with Standardized Parenteral Nutrition Solutions for Very Preterm Infants. Nutrients 2022; 14:3912. [PMID: 36235563 PMCID: PMC9572167 DOI: 10.3390/nu14193912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/09/2022] [Accepted: 09/10/2022] [Indexed: 11/16/2022] Open
Abstract
Very preterm infants are at high risk for suboptimal nutrition in the first weeks of life leading to insufficient weight gain and complications arising from metabolic imbalances such as insufficient bone mineral accretion. We investigated the use of a novel set of standardized parenteral nutrition (PN; MUC PREPARE) solutions regarding improving nutritional intake, accelerating termination of parenteral feeding, and positively affecting growth in comparison to individually prescribed and compounded PN solutions. We studied the effect of MUC PREPARE on macro- and micronutrient intake, metabolism, and growth in 58 very preterm infants and compared results to a historic reference group of 58 very preterm infants matched for clinical characteristics. Infants receiving MUC PREPARE demonstrated improved macro- and micronutrient intake resulting in balanced electrolyte levels and stable metabolomic profiles. Subsequently, improved energy supply was associated with up to 1.5 weeks earlier termination of parenteral feeding, while simultaneously reaching up to 1.9 times higher weight gain at day 28 in extremely immature infants (<27 GA weeks) as well as overall improved growth at 2 years of age for all infants. The use of the new standardized PN solution MUC PREPARE improved nutritional supply and short- and long-term growth and reduced PN duration in very preterm infants and is considered a superior therapeutic strategy.
Collapse
Affiliation(s)
- Alida Kindt
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Oberschleißheim, Germany
- Metabolomics and Analytics Center, Leiden Academic Centre for Drug Research, Leiden University, 2333 AL Leiden, The Netherlands
| | - Yvonne Kraus
- Center for Comprehensive Developmental Care (CDeCLMU), Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr von Hauner Children's Hospital, Munich University Hospital, Ludwig Maximilians University, 80336 Munich, Germany
- Comprehensive Pneumology Center, Helmholtz Zentrum München, Member of the German Lung Research Center (DZL), 81377 Munich, Germany
| | - David Rasp
- Metabolomics and Analytics Center, Leiden Academic Centre for Drug Research, Leiden University, 2333 AL Leiden, The Netherlands
| | - Kai M. Foerster
- Comprehensive Pneumology Center, Helmholtz Zentrum München, Member of the German Lung Research Center (DZL), 81377 Munich, Germany
- Department of Neonatology, Perinatal Center, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians University, 80337 Munich, Germany
| | - Narges Ahmidi
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Oberschleißheim, Germany
| | - Andreas W. Flemmer
- Department of Neonatology, Perinatal Center, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians University, 80337 Munich, Germany
| | - Susanne Herber-Jonat
- Department of Neonatology, Perinatal Center, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians University, 80337 Munich, Germany
| | - Florian Heinen
- Center for Comprehensive Developmental Care (CDeCLMU), Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr von Hauner Children's Hospital, Munich University Hospital, Ludwig Maximilians University, 80336 Munich, Germany
| | - Heike Weigand
- Center for Comprehensive Developmental Care (CDeCLMU), Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr von Hauner Children's Hospital, Munich University Hospital, Ludwig Maximilians University, 80336 Munich, Germany
| | - Thomas Hankemeier
- Metabolomics and Analytics Center, Leiden Academic Centre for Drug Research, Leiden University, 2333 AL Leiden, The Netherlands
| | - Berthold Koletzko
- Division of Metabolic and Nutritional Medicine, Department of Paediatrics, Dr. von Hauner Children’s Hospital University Hospital, Ludwig-Maximilians University, 81377 Munich, Germany
| | - Jan Krumsiek
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Oberschleißheim, Germany
- Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Juergen Babl
- Pharmacy of the University Hospital, Ludwig-Maximilians University, 81377 Munich, Germany
| | - Anne Hilgendorff
- Center for Comprehensive Developmental Care (CDeCLMU), Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr von Hauner Children's Hospital, Munich University Hospital, Ludwig Maximilians University, 80336 Munich, Germany
- Comprehensive Pneumology Center, Helmholtz Zentrum München, Member of the German Lung Research Center (DZL), 81377 Munich, Germany
- Department of Neonatology, Perinatal Center, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians University, 80337 Munich, Germany
| |
Collapse
|
9
|
Dassios T, Williams EE, Hickey A, Bunce C, Greenough A. Bronchopulmonary dysplasia and postnatal growth following extremely preterm birth. Arch Dis Child Fetal Neonatal Ed 2021; 106:386-391. [PMID: 33334820 DOI: 10.1136/archdischild-2020-320816] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/27/2020] [Accepted: 12/01/2020] [Indexed: 11/04/2022]
Abstract
OBJECTIVES To report the current incidence of bronchopulmonary dysplasia (BPD) and to compare changes in weight and head circumference between infants who developed BPD and infants who did not. DESIGN Retrospective, whole-population study. SETTING All neonatal units in England between 2014 and 2018. PATIENTS All liveborn infants born <28 completed weeks of gestation. INTERVENTIONS The change in weight z-score (ΔWz) was calculated by subtracting the birthweight z-score from the weight z-score at 36 weeks postmenstrual age (PMA) and at discharge. The change in head circumference z-score (ΔHz) was calculated by subtracting the birth head circumference z-score from the head circumference z-score at discharge. MAIN OUTCOME MEASURE BPD was defined as the need for any respiratory support at 36 weeks PMA. RESULTS 11 806 infants were included in the analysis. The incidence of BPD was 57.5%, and 18.9% of the infants died before 36 weeks PMA. The median (IQR) ΔWz from birth to 36 weeks PMA was significantly smaller in infants who developed BPD (-0.69 (-1.28 to -0.14), n=6105) than in those who did not develop BPD (-0.89 (-1.40 to -0.33), n=2390; adjusted p<0.001). The median (IQR) ΔHz from birth to discharge was significantly smaller in infants who developed BPD (-0.33 (-1.69 to 0.71)) than in those who did not develop BPD (-0.61 (-1.85 to 0.35); adjusted p<0.001). CONCLUSIONS Postnatal growth was better in infants diagnosed with BPD compared with infants without BPD possibly due to more aggressive nutrition strategies.
Collapse
Affiliation(s)
- Theodore Dassios
- Neonatal Intensive Care Unit, King's College Hospital NHS Foundation Trust, London, UK .,Department of Women and Children's Health, School of Life Course Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Emma E Williams
- Department of Women and Children's Health, School of Life Course Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Ann Hickey
- Neonatal Intensive Care Centre, King's College Hospital NHS Foundation Trust, London, UK
| | - Catey Bunce
- School of Population Health and Environmental Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Anne Greenough
- Department of Women and Children's Health, School of Life Course Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK.,Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| |
Collapse
|
10
|
Strobel KM, Romero T, Kramer K, Fernandez E, Rottkamp C, Uy C, Keller R, Moyer L, Poulain F, Kim JH, DeUgarte DA, Calkins KL. Growth Failure Prevalence in Neonates with Gastroschisis : A Statewide Cohort Study. J Pediatr 2021; 233:112-118.e3. [PMID: 33647253 PMCID: PMC8154735 DOI: 10.1016/j.jpeds.2021.02.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVES To perform a multicenter study to assess growth failure in hospitalized infants with gastroschisis. STUDY DESIGN This study included neonates with gastroschisis within sites in the University of California Fetal Consortium. The study's primary outcome was growth failure at hospital discharge, defined as a weight or length z score decrease >0.8 from birth. Regression analysis was performed to assess changes in z scores over time. RESULTS Among 125 infants with gastroschisis, the median gestational age was 37 weeks (IQR 35-37). Length of stay was 32 days (23-60); 55% developed weight or length growth failure at discharge (28% had weight growth failure, 42% had length growth failure, and 15% had both weight and length growth failure). Weight and length z scores at 14 days, 30 days, and discharge were less than birth (P < .01 for all). Weight and length z scores declined from birth to 30 days (-0.10 and -0.11 z score units/week, respectively, P < .001). Length growth failure at discharge was associated with weight and length z score changes over time (P < .05 for both). Lower gestational age was associated with weight growth failure (OR 0.70 for each gestational age week, 95% CI 0.55-0.89, P = .004). CONCLUSIONS Growth failure, in particular linear growth failure, is common in infants with gastroschisis. These data suggest the need to improve nutritional management in these infants.
Collapse
Affiliation(s)
- Katie M Strobel
- Division of Neonatology and Developmental Biology, Department of Pediatrics, University of California Los Angeles, Los Angeles, CA.
| | - Tahmineh Romero
- Division of General Internal Medicine and Health Services Research, Department of Medicine Statistics Core, University of California Los Angeles, Los Angeles, CA
| | - Katelin Kramer
- Division of Neonatology, Department of Pediatrics, University of California San Francisco, San Francisco, CA
| | - Erika Fernandez
- Division of Neonatology, Department of Pediatrics, University of California San Diego, San Diego, CA
| | - Catherine Rottkamp
- Division of Neonatology, Department of Pediatrics, University of California Davis, Davis, CA
| | - Cherry Uy
- Division of Neonatal/Perinatal Medicine, Department of Pediatrics, University of California Irvine, Irvine, CA
| | - Roberta Keller
- Division of Neonatology, Department of Pediatrics, University of California San Francisco, San Francisco, CA
| | - Laurel Moyer
- Division of Neonatology, Rady Children's Hospital, San Diego, CA
| | - Francis Poulain
- Division of Neonatology, Department of Pediatrics, University of California Davis, Davis, CA
| | - Jae H Kim
- Division of Neonatology, Department of Pediatrics, University of California San Diego, San Diego, CA
| | - Daniel A DeUgarte
- Division of Pediatric Surgery, Department of Surgery, University of California Los Angeles, Los Angeles, CA
| | - Kara L Calkins
- Division of Neonatology and Developmental Biology, Department of Pediatrics, University of California Los Angeles, Los Angeles, CA
| |
Collapse
|
11
|
Joung KE, Martin CR, Cherkerzian S, Kellogg M, Belfort MB. Human Milk Hormone Intake in the First Month of Life and Physical Growth Outcomes in Preterm Infants. J Clin Endocrinol Metab 2021; 106:1793-1803. [PMID: 33544860 DOI: 10.1210/clinem/dgab001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Indexed: 12/31/2022]
Abstract
CONTEXT Human milk contains hormones that regulate metabolism. Extrauterine growth restriction remains common among preterm infants, but the effect of ingesting milk hormones on preterm infant growth is poorly understood. OBJECTIVE To quantify associations of longitudinal exposure to leptin, adiponectin, and insulin in milk with physical growth of preterm infants. DESIGN/METHODS In 50 preterm neonates (median gestational age 29.4 weeks), we sampled maternal milk on day-of-life 7, 14, 21, and 28 and measured hormone levels in whole milk by ELISA. Milk leptin levels were available for a subset of 18 infants. We calculated milk hormone doses by multiplying the hormone level by the milk volume ingested on each day and estimated the area under the curve (AUC) to reflect longitudinal exposure. We analyzed associations of milk hormone exposure with growth outcomes in generalized estimated equations. MAIN OUTCOME MEASURES Weight gain velocity and z-scores in weight, length, head circumference, and body mass index at 36 weeks' postmenstrual age (PMA). RESULTS Higher leptin intake was associated with greater weight gain (2.17g/kg/day [95% CI, 1.31, 3.02]) and weight z-score at 36 weeks' PMA (0.30 [0.08, 0.53] higher z-score per tertile). Higher adiponectin intake was associated with greater length z-score (0.41 [0.13, 0.69]), however, this association was nullified after adjustment of protein and calorie intake. Higher adiponectin was associated with smaller head circumference z-score (-0.36 [-0.64, -0.07]). Insulin was not associated with growth outcomes. CONCLUSIONS Milk leptin and adiponectin exposures may affect growth of preterm infants. The long-term effects of milk hormones warrant further investigation.
Collapse
Affiliation(s)
- Kyoung Eun Joung
- Division of Neonatology and Newborn Medicine, Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Camilia R Martin
- Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Sara Cherkerzian
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Mark Kellogg
- Department of Laboratory Medicine, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Mandy Brown Belfort
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
12
|
Abstract
Objective A neonatal nutritional risk screening tool (NNRST) was developed by using Delphi and analytic hierarchy processes in China. We verified the accuracy of this tool and analysed whether it effectively screened neonates with nutritional risk. Design Prospective validation study. Setting and participants In total, 338 neonates who were admitted to the neonatal unit of Children’s Hospital of Chongqing Medical University from May–July 2016 completed the study. Nutritional risk screening and length and head circumference measurements were performed weekly. Weight was measured every morning, and other relevant clinical data were recorded during hospitalisation. Main outcome measures We evaluated the sensitivity, specificity, validity, reliability, and positive and negative predictive value of the screening tool. Various characteristics of neonates in different risk groups were analysed to determine the rationality of the nutritional risk classification. Results The sensitivity, specificity, and positive and negative predictive values were 85.11%, 91.07%, 60.61% and 97.43%, respectively. The criterion validity was texted by the Spearman correlation analysis (r=0.530) and independent samples non-parametric tests (p=0.000). The content validity (Spearman correlation coefficient) was 0.321–0.735. The inter-rater reliability (kappa value) was 0.890. Among the neonatal clinical indicators, gestational age, birth weight, length, admission head circumference, admission albumin, admission total proteins, discharge weight, discharge length and head circumference decreased with increasing nutrition risk level; the length of stay and the rate of parenteral nutrition support increased with increasing nutrition risk level. In the comparison of complications during hospitalisation, the incidence of necrotising enterocolitis and congenital gastrointestinal malformation increased with increasing nutrition risk level. Conclusion The validation results for the NNRST are reliable. The tool can be used to preliminarily determine the degree of neonatal nutritional risk, but its predictive value needs to be determined in future large-sample studies. Trial registration number ChiCTR2000033743.
Collapse
|
13
|
Chiavaroli V, Derraik JGB, Jayasinghe TN, Rodrigues RO, Biggs JB, Battin M, Hofman PL, O'Sullivan JM, Cutfield WS. Lower insulin sensitivity remains a feature of children born very preterm. Pediatr Diabetes 2021; 22:161-167. [PMID: 33084185 DOI: 10.1111/pedi.13140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/24/2020] [Accepted: 10/15/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The first report of children born very preterm (<32 weeks of gestation) having insulin resistance was made 16 years ago. However, neonatal care has improved since. Thus, we aimed to assess whether children born very preterm still have lower insulin sensitivity than term controls. METHODS Participants were prepubertal children aged 5 to 11 years born very preterm (<32 weeks of gestation; n = 51; 61% boys) or at term (37-41 weeks; n = 50; 62% boys). Frequently sampled intravenous glucose tolerance tests were performed, and insulin sensitivity was calculated using Bergman's minimal model. Additional clinical assessments included anthropometry, body composition using whole-body dual-energy X-ray absorptiometry scans, clinic blood pressure, and 24-hour ambulatory blood pressure monitoring. RESULTS Children born very preterm were 0.69 standard deviation score (SDS) lighter (P < .001), 0.53 SDS shorter (P = .003), and had body mass index 0.57 SDS lower (P = .003) than children born at term. Notably, children born very preterm had insulin sensitivity that was 25% lower than term controls (9.4 vs 12.6 × 10-4 minutes-1 ·[mU/L]; P = .001). Other parameters of glucose metabolism, including fasting insulin levels, were similar in the two groups. The awake systolic blood pressure (from 24-hour monitoring) tended to be 3.1 mm Hg higher on average in children born very preterm (P = .054), while the clinic systolic blood pressure was 5.4 mm Hg higher (P = .002). CONCLUSIONS Lower insulin sensitivity remains a feature of children born very preterm, despite improvements in neonatal intensive care. As reported in our original study, our findings suggest the defect in insulin action in prepubertal children born very pretermis primarily peripheral and not hepatic.
Collapse
Affiliation(s)
- Valentina Chiavaroli
- Liggins Institute, University of Auckland, Auckland, New Zealand.,Neonatal Intensive Care Unit, Pescara Public Hospital, Pescara, Italy
| | - José G B Derraik
- Liggins Institute, University of Auckland, Auckland, New Zealand.,A Better Start - National Science Challenge, University of Auckland, Auckland, New Zealand.,Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden.,Endocrinology Department, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | | | | | - Janene B Biggs
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Malcolm Battin
- Newborn Services, Auckland City Hospital, Auckland, New Zealand
| | - Paul L Hofman
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | - Wayne S Cutfield
- Liggins Institute, University of Auckland, Auckland, New Zealand.,A Better Start - National Science Challenge, University of Auckland, Auckland, New Zealand
| |
Collapse
|
14
|
Salas AA, Travers CP, Jerome ML, Chandler-Laney P, Carlo WA. Percent Body Fat Content Measured by Plethysmography in Infants Randomized to High- or Usual-Volume Feeding after Very Preterm Birth. J Pediatr 2021; 230:251-254.e3. [PMID: 33248115 PMCID: PMC7914146 DOI: 10.1016/j.jpeds.2020.11.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/21/2022]
Abstract
We measured percent body fat by air-displacement plethysmography in 86 infants born at <32 weeks of gestation randomized to receive either high-volume (180-200 mL/kg/day) or usual volume feeding (140-160 mL/kg/day). High-volume feeding increased percent body fat by ≤2% at 36 weeks of postmenstrual age (within a predefined range of equivalence). TRIAL REGISTRATION: ClincialTrials.gov: NCT02377050.
Collapse
Affiliation(s)
- Ariel A. Salas
- Department of Pediatrics, School of Medicine, University of
Alabama at Birmingham, Birmingham, AL, USA
| | - Colm P. Travers
- Department of Pediatrics, School of Medicine, University of
Alabama at Birmingham, Birmingham, AL, USA
| | - Maggie L. Jerome
- Department of Nutrition Sciences, School of Health
Professions, University of Alabama at Birmingham. Birmingham, AL, USA
| | - Paula Chandler-Laney
- Department of Nutrition Sciences, School of Health
Professions, University of Alabama at Birmingham. Birmingham, AL, USA
| | - Waldemar A. Carlo
- Department of Pediatrics, School of Medicine, University of
Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
15
|
Salas AA, Jerome ML, Chandler-Laney P, Ambalavanan N, Carlo WA. Serial assessment of fat and fat-free mass accretion in very preterm infants: a randomized trial. Pediatr Res 2020; 88:733-738. [PMID: 32634820 PMCID: PMC7581604 DOI: 10.1038/s41390-020-1052-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/26/2020] [Accepted: 06/26/2020] [Indexed: 12/03/2022]
Abstract
BACKGROUND Clinicians could modify dietary interventions during early infancy by monitoring fat and fat-free mass accretion in very preterm infants. METHODS Preterm infants were randomly assigned to either having reports on infant body composition available to the clinicians caring for them (intervention group) or not having reports available (control group). All infants underwent serial assessments of body composition by air-displacement plethysmography before 32 weeks of postmenstrual age (PMA) and at 36 weeks PMA. The primary outcome was percent body fat (%BF) at 3 months of corrected age (CA). RESULTS Fifty infants were randomized (median gestational age: 30 weeks; mean ± SD birth weight: 1387 ± 283 g). The mean %BF increased from 7 ± 4 before 32 weeks PMA to 20 ± 5 at 3 months CA. The differences in mean %BF between the intervention group and the control group were not statistically significant at 36 weeks PMA (14.5 vs. 13.6) or 3 months CA (20.8 vs. 19.4). Feeding practices and anthropometric measurements during hospitalization did not differ between groups. CONCLUSIONS Serial assessments of body composition in both intervention and control groups showed consistent increments in %BF. However, providing this information to clinicians did not influence nutritional practices or growth. IMPACT Serial assessments of body composition in preterm infants at 32 and 36 weeks postmenstrual age show consistent increments in % body fat up to 3 months of corrected age. However, providing this information to the clinician did not influence nutritional practices or growth.
Collapse
Affiliation(s)
- Ariel A Salas
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, 35249, USA.
| | - Maggie L Jerome
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, 35249, USA
| | - Paula Chandler-Laney
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, 35249, USA
| | | | - Waldemar A Carlo
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, 35249, USA
| |
Collapse
|
16
|
Ma J, Shen F, Chen L, Wu H, Huang Y, Fan Z, Hou R, Yue B, Zhang X. Gene expression profiles during postnatal development of the liver and pancreas in giant pandas. Aging (Albany NY) 2020; 12:15705-15729. [PMID: 32805731 PMCID: PMC7467380 DOI: 10.18632/aging.103783] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/14/2020] [Indexed: 01/07/2023]
Abstract
Giant pandas are unique Carnivora with a strict bamboo diet. To investigate the molecular mechanism of giant panda nutrient metabolism from newborn to adult, the gene expression profiles of giant panda liver and pancreas tissues collected from three important feeding stages were investigated using RNA-seq. We found a total of 3,211 hepatic and 3,343 pancreatic differentially expressed genes (DEGs) from three comparisons between suckling and no feeding, adult and no feeding, and adult and suckling groups. Few differences in gene-expression profiles were exhibited between no feeding and suckling groups in both tissues. GO and KEGG analyses were performed to further understand the biological functions of the DEGs. In both the liver and pancreas, genes related mainly to cell cycle processes were highly up-regulated in newborn samples whereas genes related to metabolism and immunity were up-regulated in adult giant pandas. The high expression of metabolism-related genes in adult samples probably helps to fulfill the metabolic function requirements of the liver and pancreas. In contrast, several vital genes involved in cholesterol metabolism and protein digestion and absorption were over-expressed in newborn samples. This may indicate the importance of cholesterol metabolism and protein digestion and absorption processes in giant panda infancy.
Collapse
Affiliation(s)
- Jinnan Ma
- Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China
| | - Fujun Shen
- The Sichuan Key Laboratory for Conservation Biology of Endangered Wildlife, Chengdu Research Base of Giant Panda Breeding, Chengdu 610081, China
| | - Lei Chen
- Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu 610065, China
| | - Honglin Wu
- China Conservation and Research Center for the Giant Panda, Wolong, Sichuan 623006, China
| | - Yan Huang
- China Conservation and Research Center for the Giant Panda, Wolong, Sichuan 623006, China
| | - Zhenxin Fan
- Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China.,Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu 610065, China
| | - Rong Hou
- The Sichuan Key Laboratory for Conservation Biology of Endangered Wildlife, Chengdu Research Base of Giant Panda Breeding, Chengdu 610081, China
| | - Bisong Yue
- Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China.,Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu 610065, China
| | - Xiuyue Zhang
- Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China.,Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu 610065, China
| |
Collapse
|
17
|
Colombo J, Gustafson KM, Carlson SE. Critical and Sensitive Periods in Development and Nutrition. ANNALS OF NUTRITION AND METABOLISM 2020; 75 Suppl 1:34-42. [PMID: 32554960 DOI: 10.1159/000508053] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/16/2020] [Indexed: 12/14/2022]
Abstract
Critical or sensitive periods in the life of an organism during which certain experiences or conditions may exert disproportionate influence (either for harm or benefit) on long-term developmental outcomes have been the subject of investigation for over a century. This chapter reviews research in the context of the development of social preferences and sensory systems, with a summary of the criteria for defining such a period and the evidence necessary to establish its existence. The notion of nutritional programming, central to the Barker/Developmental Origins hypotheses of health and disease, represents a variant of the critical/sensitive period concept. It is implicit in these hypotheses that the fetal period is a time during which metabolic and physiological systems are malleable and thus susceptible to either insult or enhancement by nutrient intake. Evidence for critical/sensitive periods or nutritional programming requires a systematic manipulation of the age at which nutritional conditions or supplements are implemented. While common in research using animal models, the approach is difficult to establish in epidemiological studies and virtually nonexistent in human clinical trials. Future work seeking to establish definitive evidence for critical/sensitive periods or programming may be advanced by harmonized outcome measures in experimental trials across which the timing, duration, and dose of nutrients is varied.
Collapse
Affiliation(s)
- John Colombo
- Department of Psychology and Schiefelbusch Institute for Life Span Studies, University of Kansas, Lawrence, Kansas, USA,
| | - Kathleen M Gustafson
- Hoglund Brain Imaging Center, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Susan E Carlson
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
18
|
Plasma Adipokines Profile in Prepubertal Children with a History of Prematurity or Extrauterine Growth Restriction. Nutrients 2020; 12:nu12041201. [PMID: 32344627 PMCID: PMC7231070 DOI: 10.3390/nu12041201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/15/2020] [Accepted: 04/22/2020] [Indexed: 12/27/2022] Open
Abstract
Adipose tissue programming could be developed in very preterm infants with extrauterine growth restriction (EUGR), with an adverse impact on long-term metabolic status, as was studied in intrauterine growth restriction patterns. The aim of this cohort study was to evaluate the difference in levels of plasma adipokines in children with a history of EUGR. A total of 211 school age prepubertal children were examined: 38 with a history of prematurity and EUGR (EUGR), 50 with a history of prematurity with adequate growth (PREM), and 123 healthy children born at term. Anthropometric parameters, blood pressure, metabolic markers and adipokines (adiponectin, resistin, leptin) were measured. Children with a history of EUGR showed lower values of adiponectin (μg/mL) compared with the other two groups: (EUGR: 10.6 vs. PREM: 17.7, p < 0.001; vs. CONTROL: 25.7, p = 0.004) and higher levels of resistin (ng/mL) (EUGR: 19.2 vs. PREM: 16.3, p =0.007; vs. CONTROL: 7.1, p < 0.001. The PREM group showed the highest values of leptin (ng/mL), compared with the others: PREM: 4.9 vs. EUGR: 2.1, p = 0.048; vs. CONTROL: 3.2, p = 0.029). In conclusion, EUGR in premature children could lead to a distinctive adipokines profile, likely associated with an early programming of the adipose tissue, and likely to increase the risk of adverse health outcomes later in life.
Collapse
|
19
|
Use of the Lean Manufacturing Principles to Improve Total Parenteral Nutrition Logistics and Clinical Outcomes in the Neonatal Patient Population. Pediatr Qual Saf 2020; 4:e233. [PMID: 32010859 PMCID: PMC6946236 DOI: 10.1097/pq9.0000000000000233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 10/16/2019] [Indexed: 11/28/2022] Open
Abstract
Supplemental Digital Content is available in the text. Total parenteral nutrition (TPN) is one of the most frequently used pharmaceuticals administered to patients in our Neonatal Intensive Care Unit (NICU). Initially, the total interdepartmental processing time (ordering, manufacturing, and delivery between NICU and Pharmacy) averaged 15.2 hours. Inefficiencies in this process only allowed TPN to infuse 8.8 hours on average before labs were collected the next morning. Given the short administration-to-laboratory collection time, we hypothesized that laboratory samples would not adequately reflect the effect of the current TPN infusion. Furthermore, clinicians would be making decisions based on suboptimal data and ultimately nourish this patient population inadequately.
Collapse
|
20
|
Martin-Harris B, Carson KA, Pinto JM, Lefton-Greif MA. BaByVFSSImP © A Novel Measurement Tool for Videofluoroscopic Assessment of Swallowing Impairment in Bottle-Fed Babies: Establishing a Standard. Dysphagia 2020; 35:90-98. [PMID: 30955137 PMCID: PMC6778717 DOI: 10.1007/s00455-019-10008-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 03/28/2019] [Indexed: 10/27/2022]
Abstract
AIM This investigation tested the construct validity of the first standardized assessment tool, the BaByVFSS Impairment Profile, (BaByVFSSImP©), developed for the quantification of swallowing observations made from videofluoroscopic swallow studies (VFSS) in bottle-fed babies. METHOD Construct validity of the measures was tested using descriptive methods and confirmatory factor analysis (CFA) of swallowing scores obtained from a cohort of bottle-fed babies (median age 3 months 1 day, interquartile range 1 month 4 days-7 months 4 days) sequentially referred for VFSS based on clinical signs, symptoms, or risk factors associated with dysphagia and/or aspiration. Main outcome measures were emergence of functional domains derived from swallowing component impairment scores. RESULTS Confirmatory factor analysis resulted in 21 significant components (factor loadings ≥ 0.5) grouping into five functional domains labeled for common contribution to overall swallowing function. The tool was organized into the BaByVFSSImP. Clinical relevance was explored using correlational analyses between domain scores, maximum penetration/aspiration scores, feeding status, and caregiver burden. INTERPRETATION Quantification of physiologic swallowing impairment captured by BaByVFSSImP holds promise for identification of physiologically based targets for intervention, clinical decisions regarding enteral feeding, and tracking the trajectory of swallowing impairment throughout development in young children.
Collapse
Affiliation(s)
- Bonnie Martin-Harris
- Department of Communication Sciences and Disorders, Department of Otolaryngology Head and Neck Surgery, Department of Radiation Oncology, School of Communication, Feinberg School of Medicine, Northwestern University, 2240 Campus Drive, Evanston, IL, 60208, USA.
- Department of Otolaryngology Head and Neck Surgery, Evelyn Trammell Institute for Voice and Swallowing, College of Medicine, Medical University of South Carolina, 135 Rutledge Avenue, MSC 550, Charleston, SC, 29425, USA.
- Research Service, Edward Hines, Jr. VA Hospital, 5000 South 5th Avenue, Hines, IL, 60141, USA.
| | - Kathryn A Carson
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Welch Center for Prevention, Epidemiology and Clinical Research, 2024 East Monument Street, Suite 2-500, Baltimore, MD, 21287, USA
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Jeanne M Pinto
- Eudowood Division of Pediatric Respiratory Sciences, Department of Pediatrics, The Johns Hopkins University School of Medicine, David M. Rubenstein Building, Suite 3070, 200 North Wolfe Street, Baltimore, MD, 21287, USA
| | - Maureen A Lefton-Greif
- Eudowood Division of Pediatric Respiratory Sciences, Department of Pediatrics, The Johns Hopkins University School of Medicine, David M. Rubenstein Building, Suite 3017, 200 North Wolfe Street, Baltimore, MD, 21287, USA
- Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Physical Medicine and Rehabilitation, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| |
Collapse
|
21
|
Early energy restriction in premature infants and bronchopulmonary dysplasia: a cohort study. Br J Nutr 2020; 123:1024-1031. [PMID: 31964427 DOI: 10.1017/s0007114520000240] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is a multifactor pathology. Animal studies and cohort studies suggest that poor nutrient intake after birth increases the risk of BPD. The objective of the present study was to determine the existence of association between BPD in very low birth weight (VLBW) and energy intake during the first week of life. We recorded in a retrospective cohort study the intake of enteral and parenteral macronutrients during this period by examining the nutritional and clinical history of 450 VLBW newborns admitted to the neonatal intensive care unit. After applying the relevant exclusion criteria, data for 389 VLBW infants were analysed, of whom 159 developed some degree of BPD. Among the newborns with BPD, energy and lipid intake was significantly lower and fluid intake was significantly higher. The energy intake for the 25th percentile in the group without BPD was 1778·2 kJ/kg during the first week of life. An energy intake <1778·2 kJ/kg in this period was associated with a 2-fold increase in the adjusted risk of BPD (OR 2·63, 95 % CI 1·30, 5·34). The early nutrition and the increase of energy intake in the first week of life are associated in our sample with a lower risk of BPD developing.
Collapse
|
22
|
Maternal high-fat diet induces sex-specific endocannabinoid system changes in newborn rats and programs adiposity, energy expenditure and food preference in adulthood. J Nutr Biochem 2018; 51:56-68. [DOI: 10.1016/j.jnutbio.2017.09.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/28/2017] [Indexed: 12/17/2022]
|
23
|
Stimulation of liver IGF-1 expression promotes peak bone mass achievement in growing rats: a study with pomegranate seed oil. J Nutr Biochem 2017; 52:18-26. [PMID: 29121593 DOI: 10.1016/j.jnutbio.2017.09.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 08/14/2017] [Accepted: 09/28/2017] [Indexed: 01/08/2023]
Abstract
Peak bone mass (PBM) achieved at adulthood is a strong determinant of future onset of osteoporosis, and maximizing it is one of the strategies to combat the disease. Recently, pomegranate seed oil (PSO) has been shown to have bone-sparing effect in ovariectomized mice. However, its effect on growing skeleton and its molecular mechanism remain unclear. In the present study, we evaluated the effect of PSO on PBM in growing rats and associated mechanism of action. PSO was given at various doses to 21-day-old growing rats for 90 days by oral gavage. The changes in bone parameters were assessed by micro-computed tomography and histology. Enzyme-linked immunosorbent assay was performed to analyze the levels of serum insulin-like growth factor type 1 (IGF-1). Western blotting from bone and liver tissues was done. Chromatin immunoprecipitation assay was performed to study the histone acetylation levels at IGF-1 gene. The results of the study show that PSO treatment significantly increases bone length, bone formation rate, biomechanical parameters, bone mineral density and bone microarchitecture along with enhancing muscle and brown fat mass. This effect was due to the increased serum levels of IGF-1 and stimulation of its signaling in the bones. Studies focusing on acetylation of histones in the liver, the major site of IGF-1 synthesis, showed enrichment of acetylated H3K9 and H3K14 at IGF-1 gene promoter and body. Further, the increased acetylation at H3K9 and H3K14 was associated with a reduced HDAC1 protein level. Together, our data suggest that PSO promotes the PBM achievement via increased IGF-1 expression in liver and IGF-1 signaling in bone.
Collapse
|
24
|
Neonatal milk supplementation in lambs has persistent effects on growth and metabolic function that differ by sex and gestational age. Br J Nutr 2016; 116:1912-1925. [DOI: 10.1017/s0007114516004013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
AbstractThe perinatal environment has a major influence on long-term health and disease risk. Preterm birth alters early-life environment and is associated with altered metabolic function in adulthood. Whether preterm birthper seor the early nutritional interventions used to support growth in preterm infants underpins this association is unknown. Lambs born preterm, following dexamethasone induction of labour, or spontaneously at term were randomised to receive nutrient supplementation, analogous to the milk fortifier used clinically or water as a control for the first 2 weeks after birth. Thereafter, nutrition was not different between groups. Growth was monitored, and the glucose–insulin axis function was assessed in juvenile (4 months) and adult life (14 months). Early nutrition influenced adult metabolic function and body composition to a greater extent than preterm birth. In supplemented females, arginine-stimulated insulin secretion was increased in preterm but reduced in term-born juveniles compared with controls (repeated-measures ANOVAP<0·01). In supplemented preterm males, adult weight, ponderal index (PI) and fasting insulin concentrations were elevated compared with preterm controls (weight, 75 (sem3)v. 69 (sem2) kg; PI, 48·0 (sem2·1)v. 43·7 (sem1·7) kg/m3; fasting insulin, 0·19 (sem0·02)v. 0·10 (sem0·02) ng/ml). Conversely, supplemented term-born males had reduced adult weight, PI and fasting insulin concentrations compared with term-born controls (weight, 64 (sem2)v. 70 (sem2) kg; PI, 44·4 (sem1·8)v. 48·2 (sem1·7) kg/m3; fasting insulin, 0·09 (sem0·02)v. 0·14 (sem0·02) ng/ml; all group×supplement interactionsP<0·05). Adult metabolic health may reflect both gestational age at birth and early nutrition. Human studies are urgently needed to investigate the adult sex-specific health implications of neonatal nutritional strategies.
Collapse
|
25
|
Li Z, Valentine CJ, Wang D, Wang L, Dong M. Comparison of enteral feeding of preterm infants between two hospitals in China and United States. J Matern Fetal Neonatal Med 2016; 30:121-125. [PMID: 27345030 DOI: 10.3109/14767058.2016.1163681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIM To compare the details of preterm infants enteral feeding between the two hospitals in China and in the United States, and to analyze the reason of the differences. METHODS A retrospective cohort study was conducted. Infants < 32 weeks were enrolled from Cincinnati University Hospital (CUH) during January 2011 to January 2012 and Peking Union Medical College Hospital (PUMCH) during January 2011 to May 2012. Basic data and enteral feeding data of the two groups were compared. RESULTS Eighty-two infants in CUH group and 74 infants in PUMCH group were enrolled, infants in CUH group were much smaller than PUMCH group (gestational age (29.1 ± 2.0) versus (30.6 ± 1.3) weeks, p = 0.000, birth weight (1204 ± 328) versus (1406 ± 320) g, p = 0.000). Significantly more infants in CUH group received human milk as the first enteral feeding (78/82 versus 7/74, p = 0.000). Human milk feeding rate in first 28 days in CUH group was much higher (77/82 versus 7/74, p = 0.000). The initial milk volume, and the milk volume on the 7th, 14th, 21st and 27th day of CUH group were significant larger [(15.9 versus 9.3 ml/kg·d, p = 0.000), (79.8 versus 35.2 ml/kg·d, p = 0.000), (133.2 versus 76.4 ml/kg·d, p = 0.000), (140.6 versus 108.6 ml/kg·d, p = 0.000), (142.2 versus 121.5 ml/kg·d, p = 0.002)]. CUH group achieved full enteral feeding sooner (12.0 versus 22.4 d, p = 0.000). CONCLUSION Preterm infants achieved full enteral feeding sooner at CUH compared to PUMCH. Human milk feeding may improve enteral feeding tolerance. We need more aggressive enteral feeding proposal in PUMCH.
Collapse
Affiliation(s)
- Zhenghong Li
- a Department of Pediatrics , Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences , Beijing , China and
| | - Christina J Valentine
- b Section of Neonatology Perinatal & Pulmonary Biology, Cincinnati Children's Hospital Medical Center, The University of Cincinnati , Cincinnati , OH , USA
| | - Danhua Wang
- a Department of Pediatrics , Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences , Beijing , China and
| | - Lin Wang
- a Department of Pediatrics , Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences , Beijing , China and
| | - Mei Dong
- a Department of Pediatrics , Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences , Beijing , China and
| |
Collapse
|
26
|
Ward WE, Kaludjerovic J, Dinsdale EC. A Mouse Model for Studying Nutritional Programming: Effects of Early Life Exposure to Soy Isoflavones on Bone and Reproductive Health. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:E488. [PMID: 27187422 PMCID: PMC4881113 DOI: 10.3390/ijerph13050488] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/03/2016] [Accepted: 05/05/2016] [Indexed: 12/17/2022]
Abstract
Over the past decade, our research group has characterized and used a mouse model to demonstrate that "nutritional programming" of bone development occurs when mice receive soy isoflavones (ISO) during the first days of life. Nutritional programming of bone development can be defined as the ability for diet during early life to set a trajectory for better or compromised bone health at adulthood. We have shown that CD-1 mice exposed to soy ISO during early neonatal life have higher bone mineral density (BMD) and greater trabecular inter-connectivity in long bones and lumbar spine at young adulthood. These skeletal sites also withstand greater forces before fracture. Because the chemical structure of ISO resembles that of 17-β-estradiol and can bind to estrogen receptors in reproductive tissues, it was prudent to expand analyses to include measures of reproductive health. This review highlights aspects of our studies in CD-1 mice to understand the early life programming effects of soy ISO on bone and reproductive health. Preclinical mouse models can provide useful data to help develop and guide the design of studies in human cohorts, which may, depending on findings and considerations of safety, lead to dietary interventions that optimize bone health.
Collapse
Affiliation(s)
- Wendy E Ward
- Department of Kinesiology, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, ON L2S 3A1, Canada.
| | - Jovana Kaludjerovic
- Department of Kinesiology, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, ON L2S 3A1, Canada.
| | - Elsa C Dinsdale
- Department of Kinesiology, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, ON L2S 3A1, Canada.
| |
Collapse
|
27
|
Strandvik B, Ntoumani E, Lundqvist-Persson C, Sabel KG. Long-chain saturated and monounsaturated fatty acids associate with development of premature infants up to 18 months of age. Prostaglandins Leukot Essent Fatty Acids 2016; 107:43-9. [PMID: 26858144 DOI: 10.1016/j.plefa.2016.01.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 12/18/2015] [Accepted: 01/12/2016] [Indexed: 10/22/2022]
Abstract
Myelination is important perinatally and highly dependent on long-chain saturated and monounsaturated fatty acids. Long-chain polyunsaturated fatty acids, nowadays often supplemented, inhibit oleic acid synthesis. Using data from a premature cohort, we studied if nervonic, lignoceric and oleic acids correlated to growth and early development up to 18 months corrected age. Small for gestational age infants had lower concentrations than infants appropriate for gestational age. Only oleic acid was negatively correlated to long-chain polyunsaturated fatty acids. Oleic and lignoceric acids correlated to social interaction at one month, and nervonic acid to mental, psychomotor and behavioral development at 6, 10 and 18 months, also when adjusted for several confounders. Negative association between oleic acid and long-chain polyunsaturated fatty acids suggests inhibition of delta-9 desaturase, and nervonic acid´s divergent correlation to lignoceric and oleic acids suggests different metabolism in neonatal period. Our results may have implications for the supplementation of premature infants.
Collapse
Affiliation(s)
- Birgitta Strandvik
- Dept. of Biosciences and Nutrition, Karolinska Institutet, NOVUM, Hälsovägen 7-9, 14183 Huddinge, Stockholm, Sweden.
| | - Eleni Ntoumani
- Dept. of Neonatology, Borås Children׳s Hospital, South Älvsborg׳s Hospital, Borås, Sweden
| | - Cristina Lundqvist-Persson
- Skaraborg Institute for Research and Development, Skövde, Sweden; Dept. of Psychology, Lund University, Lund, Sweden
| | - Karl-Göran Sabel
- Dept. of Neonatology, Borås Children׳s Hospital, South Älvsborg׳s Hospital, Borås, Sweden
| |
Collapse
|
28
|
Extrauterine growth restriction on pulmonary vascular endothelial dysfunction in adult male rats: the role of epigenetic mechanisms. J Hypertens 2016; 32:2188-98; discussion 2198. [PMID: 25105456 PMCID: PMC4222616 DOI: 10.1097/hjh.0000000000000309] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Objective: Early postnatal life is considered as a critical time window for the determination of long-term metabolic states and organ functions. Extrauterine growth restriction (EUGR) causes the development of adult-onset chronic diseases, including pulmonary hypertension. However, the effects of nutritional disadvantages during the early postnatal period on pulmonary vascular consequences in later life are not fully understood. Our study was designed to test whether epigenetics dysregulation mediates the cellular memory of this early postnatal event. Methods and results: To test this hypothesis, we isolated pulmonary vascular endothelial cells by magnetic-activated cell sorting from EUGR and control rats. A postnatal insult, nutritional restriction-induced EUGR caused development of an increased pulmonary artery pressure at 9 weeks of age in male Sprague–Dawley rats. Methyl-DNA immune precipitation chip, genome-scale mapping studies to search for differentially methylated loci between control and EUGR rats, revealed significant difference in cytosine methylation between EUGR and control rats. EUGR changes the cytosine methylation at approximately 500 loci in male rats at 9 weeks of age, preceding the development of pulmonary hypertension and these represent the candidate loci for mediating the pathogenesis of pulmonary vascular disease that occurs later in life. Gene ontology analysis on differentially methylated genes showed that hypermethylated genes in EUGR are vascular development-associated genes and hypomethylated genes in EUGR are late-differentiation-associated and signal transduction genes. We validated candidate dysregulated loci with the quantitative assays of cytosine methylation and gene expressions. Conclusion: These results demonstrate that epigenetics dysregulation is a strong mechanism for propagating the cellular memory of early postnatal events, causing changes in the expression of genes and long-term susceptibility to pulmonary hypertension, and further providing a new insight into the prevention and treatment of EUGR-related pulmonary hypertension.
Collapse
|
29
|
Postnatal high-fat diet enhances ectopic fat deposition in pigs with intrauterine growth retardation. Eur J Nutr 2015; 56:483-490. [PMID: 26707995 DOI: 10.1007/s00394-015-1093-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/26/2015] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Intrauterine growth retardation (IUGR) and postnatal nutrition are risk factors for adult metabolic syndrome. However, the influences of long-term high-fat diet (HFD) intake on ectopic fat deposition in non-adipose tissues in IUGR pigs remain unclear. The present study was to determine whether HFD consumption would enhance ectopic fat deposition in IUGR pigs. METHODS At day 28, IUGR and control pigs were fed ad libitum to either a regular diet or a HFD. Lipid store, enzymatic activities and mRNA expression of lipid metabolism-related factors in liver and semitendinosus muscle (SM) were quantified at postnatal day 178. RESULTS Feeding a HFD to IUGR pigs but not to control pigs significantly increased daily weight gain, carcass fat mass, plasma leptin level and lipid content and lipoprotein lipase (LPL) activity and mRNA abundances of LPL and peroxisome proliferator-activated receptor gamma (PPARγ) in liver and SM, but decreased daily feed intake and mRNA expression of hormone-sensitive lipase (LIPE) and carnitine palmitoyl transferase-1 (CPT-1) in liver and SM (P < 0.05). Compared with control pigs, IUGR pigs had a lower body weight but higher plasma levels of total cholesterol (TC) and insulin (P < 0.05). HFD-fed pigs exhibited greater body weight, plasma concentrations of triglyceride (TG), high-density lipoprotein cholesterol (HDL-C) and low-density lipoprotein cholesterol (LDL-C), regardless of birth weight (P < 0.05). CONCLUSION Our results suggested that IUGR increased the vulnerability of HFD-fed pigs to ectopic fat deposition via enhanced fatty acid flux toward ectopic sites and reduced lipolysis and fatty acid oxidation.
Collapse
|
30
|
Horbar JD, Ehrenkranz RA, Badger GJ, Edwards EM, Morrow KA, Soll RF, Buzas JS, Bertino E, Gagliardi L, Bellù R. Weight Growth Velocity and Postnatal Growth Failure in Infants 501 to 1500 Grams: 2000-2013. Pediatrics 2015; 136:e84-92. [PMID: 26101360 DOI: 10.1542/peds.2015-0129] [Citation(s) in RCA: 234] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Very low birth weight infants often gain weight poorly and demonstrate growth failure during the initial hospitalization. Although many of the major morbidities experienced by these infants during their initial NICU stays have decreased in recent years, it is unclear whether growth has improved. METHODS We studied 362 833 infants weighing 501 to 1500 g without major birth defects born from 2000 to 2013 and who were hospitalized for 15 to 175 days at 736 North American hospitals in the Vermont Oxford Network. Average growth velocity (GV; g/kg per day) was computed by using a 2-point exponential model on the basis of birth weight and discharge weight. Postnatal growth failure and severe postnatal growth failure were defined as a discharge weight less than the 10th and third percentiles for postmenstrual age, respectively. RESULTS From 2000 to 2013, average GV increased from 11.8 to 12.9 g/kg per day. Postnatal growth failure decreased from 64.5% to 50.3% and severe postnatal growth failure from 39.8% to 27.5%. The interquartile ranges for the hospitals participating in 2013 were as follows: GV, 12.3 to 13.4 g/kg per day; postnatal growth failure, 41.1% to 61.7%; and severe postnatal growth failure, 19.4% to 36.0%. Adjusted and unadjusted estimates were nearly identical. CONCLUSIONS For infants weighing 501 to 1500 g at birth, average GV increased and the percentage with postnatal growth failure decreased. However, in 2013, half of these infants still demonstrated postnatal growth failure and one-quarter demonstrated severe postnatal growth failure.
Collapse
Affiliation(s)
- Jeffrey D Horbar
- Vermont Oxford Network, Burlington, Vermont; Department of Pediatrics
| | | | | | - Erika M Edwards
- Vermont Oxford Network, Burlington, Vermont; Department of Mathematics and Statistics, University of Vermont, Burlington, Vermont;
| | | | - Roger F Soll
- Vermont Oxford Network, Burlington, Vermont; Department of Pediatrics
| | - Jeffrey S Buzas
- Vermont Oxford Network, Burlington, Vermont; Department of Mathematics and Statistics, University of Vermont, Burlington, Vermont
| | - Enrico Bertino
- Italian Neonatal Network, Lecco, Lido di Camaiore, and Turin, Italy
| | - Luigi Gagliardi
- Italian Neonatal Network, Lecco, Lido di Camaiore, and Turin, Italy
| | - Roberto Bellù
- Italian Neonatal Network, Lecco, Lido di Camaiore, and Turin, Italy
| |
Collapse
|
31
|
Konstantyner T, Taddei JAAC, Konstantyner TCRO, Rodrigues LC. Frequency of nutritional disorders and their risk factors among children attending 13 nurseries in São Paulo, Brazil. A cross-sectional study. SAO PAULO MED J 2015; 133:326-35. [PMID: 26517146 PMCID: PMC10876351 DOI: 10.1590/1516-3180.2014.8800711] [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: 04/23/2014] [Revised: 04/23/2014] [Accepted: 12/23/2014] [Indexed: 11/22/2022] Open
Abstract
CONTEXT AND OBJECTIVE Nutritional disorders are associated with health problems earlier in life. The objective here was to estimate the frequency of nutritional disorders and their risk factors among children. DESIGN AND SETTING Cross-sectional study in nurseries at 13 day-care centers in São Paulo, Brazil. METHODS The mothers of 482 children were interviewed, with anthropometry on these children. Children whose anthropometric indices for weight and height were greater than two standard deviations were considered to have nutritional disorders. RESULTS Children in families with lower per capita income (odds ratio [OR]: 2.25; 95% confidence interval, CI: 1.08-4.67) and who presented neonatal risk (OR 8.08; 95% CI: 2.29-28.74), had incomplete vaccinations (OR 3.44; 95% CI: 1.15-10.31) or were male (OR 3.73; 95% CI: 1.63-8.56) were more likely to be malnourished. Children in families with lower per capita income were also less likely to be overnourished (OR 0.40; 95% CI: 0.19-0.88). Children who were exclusively breastfed for less than two months (OR 2.95; 95% CI: 1.35-6.44) or who were male (OR 2.18; 95% CI: 1.02-4.65) were also at greater risk of being overnourished. Children who presented neonatal risk (OR 3.41; 95% CI: 1.04-11.23), had incomplete vaccinations (OR 3.18; 95% CI: 1.307.76), or were male (OR 2.76; 95% CI: 1.56-4.90) were more likely to have a nutritional disorder. CONCLUSIONS Nutritional disorders remain present in children attending nurseries in São Paulo. Actions should focus on boys, children who were exclusively breastfed for less than two months and those without up-to-date vaccinations.
Collapse
Affiliation(s)
- Tulio Konstantyner
- MD, MSc, PhD. Professor, Department of Health Sciences, Universidade de Santo Amaro (Unisa), São Paulo, Brazil.
| | - José Augusto Aguiar Carrazedo Taddei
- MD, MSc, PhD. Affiliated Professor, Department of Pediatrics, Escola Paulista de Medicina-Universidade Federal de São Paulo (EPM-Unifesp), São Paulo, Brazil.
| | | | - Laura Cunha Rodrigues
- MD, MSc, PhD. Head of Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom.
| |
Collapse
|
32
|
Chu A, Gozal D, Cortese R, Wang Y. Cardiovascular dysfunction in adult mice following postnatal intermittent hypoxia. Pediatr Res 2015; 77:425-33. [PMID: 25518007 DOI: 10.1038/pr.2014.197] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 09/16/2014] [Indexed: 01/13/2023]
Abstract
BACKGROUND Ex-premature infants are at higher risk for hypertension and cardiovascular disease as adults, although the mechanisms underlying such increased risks are unknown. We hypothesize that postnatal exposure to intermittent hypoxia (IH) leads to cardiovascular dysfunction in adulthood with alterations of the renin-angiotensin pathway. METHODS Neonatal mice were exposed to IH for 4 wk. At the age of 3 mo, various cardiovascular measurements were obtained. RESULTS IH-exposed mice exhibited higher systolic blood pressure, impaired baroreflex responses, and decreased heart rate variability. Furthermore, IH-exposed mice manifested evidence of endothelial dysfunction, as shown by reduced reperfusion indices after tail vessel occlusion and impaired vasodilatory responses to acetylcholine. CD31(+) endothelial cells isolated from mesenteric arteries of IH-exposed mice expressed higher levels of angiotensin-converting enzyme and reactive oxygen species; plasma angiotensin-II levels were also significantly higher in these animals. In addition, DNA methylation patterns of the Ace1 and the Agt genes in these cells were congruent with their expression patterns. CONCLUSION Our results suggest that exposures to postnatal IH alter the normal development of the renin-angiotensin system and promote the occurrence of cardiovascular dysfunction during adulthood in mice.
Collapse
Affiliation(s)
- Alison Chu
- Department of Pediatrics, University of Chicago, Chicago, Illinois
| | - David Gozal
- Department of Pediatrics, University of Chicago, Chicago, Illinois
| | - Rene Cortese
- Department of Pediatrics, University of Chicago, Chicago, Illinois
| | - Yang Wang
- Department of Pediatrics, University of Chicago, Chicago, Illinois
| |
Collapse
|
33
|
Uthaya S, Modi N. Practical preterm parenteral nutrition: systematic literature review and recommendations for practice. Early Hum Dev 2014; 90:747-53. [PMID: 25263586 DOI: 10.1016/j.earlhumdev.2014.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Current practice in relation to the prescribing, compounding and administration of parenteral nutrition for extremely preterm infants is inconsistent and based on largely historical evidence. Increasingly there are calls for more 'aggressive' nutritional interventions to prevent 'postnatal growth failure'. However the evidence base for these recommendations is weak, and there are no long-term studies examining the impact of such practices. Here we summarise the evidence for preterm parenteral nutrition interventions. We suggest principles to guide practice based on evidence from a systematic search and review of evidence to date, and recommend actions necessary to advance the understanding of this important aspect of preterm care.
Collapse
Affiliation(s)
- S Uthaya
- Imperial College London, UK; Chelsea Westminster Hospital NHS Foundation Trust, London, UK.
| | - N Modi
- Imperial College London, UK; Chelsea Westminster Hospital NHS Foundation Trust, London, UK.
| |
Collapse
|
34
|
Lawn JE, Blencowe H, Oza S, You D, Lee ACC, Waiswa P, Lalli M, Bhutta Z, Barros AJD, Christian P, Mathers C, Cousens SN. Every Newborn: progress, priorities, and potential beyond survival. Lancet 2014; 384:189-205. [PMID: 24853593 DOI: 10.1016/s0140-6736(14)60496-7] [Citation(s) in RCA: 1211] [Impact Index Per Article: 110.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In this Series paper, we review trends since the 2005 Lancet Series on Neonatal Survival to inform acceleration of progress for newborn health post-2015. On the basis of multicountry analyses and multi-stakeholder consultations, we propose national targets for 2035 of no more than 10 stillbirths per 1000 total births, and no more than 10 neonatal deaths per 1000 livebirths, compatible with the under-5 mortality targets of no more than 20 per 1000 livebirths. We also give targets for 2030. Reduction of neonatal mortality has been slower than that for maternal and child (1-59 months) mortality, slowest in the highest burden countries, especially in Africa, and reduction is even slower for stillbirth rates. Birth is the time of highest risk, when more than 40% of maternal deaths (total about 290,000) and stillbirths or neonatal deaths (5·5 million) occur every year. These deaths happen rapidly, needing a rapid response by health-care workers. The 2·9 million annual neonatal deaths worldwide are attributable to three main causes: infections (0·6 million), intrapartum conditions (0·7 million), and preterm birth complications (1·0 million). Boys have a higher biological risk of neonatal death, but girls often have a higher social risk. Small size at birth--due to preterm birth or small-for-gestational-age (SGA), or both--is the biggest risk factor for more than 80% of neonatal deaths and increases risk of post-neonatal mortality, growth failure, and adult-onset non-communicable diseases. South Asia has the highest SGA rates and sub-Saharan Africa has the highest preterm birth rates. Babies who are term SGA low birthweight (10·4 million in these regions) are at risk of stunting and adult-onset metabolic conditions. 15 million preterm births, especially of those younger than 32 weeks' gestation, are at the highest risk of neonatal death, with ongoing post-neonatal mortality risk, and important risk of long-term neurodevelopmental impairment, stunting, and non-communicable conditions. 4 million neonates annually have other life-threatening or disabling conditions including intrapartum-related brain injury, severe bacterial infections, or pathological jaundice. Half of the world's newborn babies do not get a birth certificate, and most neonatal deaths and almost all stillbirths have no death certificate. To count deaths is crucial to change them. Failure to improve birth outcomes by 2035 will result in an estimated 116 million deaths, 99 million survivors with disability or lost development potential, and millions of adults at increased risk of non-communicable diseases after low birthweight. In the post-2015 era, improvements in child survival, development, and human capital depend on ensuring a healthy start for every newborn baby--the citizens and workforce of the future.
Collapse
Affiliation(s)
- Joy E Lawn
- Centre for Maternal Reproductive & Child Health, and Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK; Saving Newborn Lives/Save the Children USA, Washington, DC, USA; Research and Evidence Division, Department for International Development, London, UK.
| | - Hannah Blencowe
- Centre for Maternal Reproductive & Child Health, and Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Shefali Oza
- Centre for Maternal Reproductive & Child Health, and Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Danzhen You
- Division of Policy and Strategy, UNICEF, New York, NY, USA
| | - Anne C C Lee
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Peter Waiswa
- Makerere University, School of Public Health, Kampala, Uganda; Division of Global Health, Karolinska Institutet, Stockholm, Sweden
| | - Marek Lalli
- Centre for Maternal Reproductive & Child Health, and Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Zulfiqar Bhutta
- Center for Global Child Health, Hospital for Sick Children, Toronto, ON, Canada; Center of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Aluisio J D Barros
- Universidade Federal de Pelotas, Pelotas, Brasil; Countdown to 2015 Equity Technical Working Group, Pelotas, Brasil
| | - Parul Christian
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Colin Mathers
- Mortality and Burden of Disease Unit, WHO, Geneva, Switzerland
| | - Simon N Cousens
- Centre for Maternal Reproductive & Child Health, and Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
35
|
Additive effects of maternal high fat diet during lactation on mouse offspring. PLoS One 2014; 9:e92805. [PMID: 24664181 PMCID: PMC3963955 DOI: 10.1371/journal.pone.0092805] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 02/26/2014] [Indexed: 12/30/2022] Open
Abstract
Recent reports indicated that nutrition in early infancy might influence later child health outcomes such as obesity and metabolic syndrome. Therefore, we examined the effects of maternal high fat diet (HFD) during lactation on the onset of a metabolic syndrome in their offspring. All offspring were cross-fostered by dams on the same or opposite diet to yield 4 groups: offspring from HFD-fed dams suckled by HFD-fed dams (OHH) and by control diet (CD)-fed dams (OHC) and CD-fed dams suckled by HFD-fed dams (OCH) and by CD-fed dams (OCC) mice. We examined several metabolic syndrome-related factors including body weight, blood pressure, glucose tolerance and adipocytokines. Mean body weights of OHH and OCH mice were significantly higher than those of OHC and OCC mice, respectively, with elevated systolic blood pressure. Moreover, OHH and OCH mice revealed significantly worse glucose tolerance compared with the OHC and OCC mice, respectively. Triglyceride and leptin levels were significantly increased and adiponectin levels were significantly reduced by the maternal HFD during lactation, with similar changes in leptin and adiponectin mRNA expression but without histone modifications in adipose tissues. In addition, maternal obesity induced by HFD during lactation increased and prolonged the leptin surge in the offspring and the gender differences of leptin surge were observed. Our data suggested that maternal HFD during lactation might have an additive effect on the onset of the metabolic syndrome in the offspring, irrespective of the nutritional status in utero through the modified leptin surge.
Collapse
|
36
|
Poore KR, Hollis LJ, Murray RJS, Warlow A, Brewin A, Fulford L, Cleal JK, Lillycrop KA, Burdge GC, Hanson MA, Green LR. Differential pathways to adult metabolic dysfunction following poor nutrition at two critical developmental periods in sheep. PLoS One 2014; 9:e90994. [PMID: 24603546 PMCID: PMC3946277 DOI: 10.1371/journal.pone.0090994] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 02/07/2014] [Indexed: 12/12/2022] Open
Abstract
Epidemiological and experimental studies suggest early nutrition has long-term effects on susceptibility to obesity, cardiovascular and metabolic diseases. Small and large animal models confirm the influence of different windows of sensitivity, from fetal to early postnatal life, on offspring phenotype. We showed previously that undernutrition in sheep either during the first month of gestation or immediately after weaning induces differential, sex-specific changes in adult metabolic and cardiovascular systems. The current study aims to determine metabolic and molecular changes that underlie differences in lipid and glucose metabolism induced by undernutrition during specific developmental periods in male and female sheep. Ewes received 100% (C) or 50% nutritional requirements (U) from 1–31 days gestation, and 100% thereafter. From weaning (12 weeks) to 25 weeks, offspring were then fed either ad libitum (CC, UC) or were undernourished (CU, UU) to reduce body weight to 85% of their individual target. From 25 weeks, all offspring were fed ad libitum. A cohort of late gestation fetuses were studied after receiving either 40% nutritional requirements (1–31 days gestation) or 50% nutritional requirements (104–127 days gestation). Post-weaning undernutrition increased in vivo insulin sensitivity, insulin receptor and glucose transporter 4 expression in muscle, and lowered hepatic methylation at the delta-like homolog 1/maternally expressed gene 3 imprinted cluster in adult females, but not males. Early gestational undernutrition induced lower hepatic expression of gluconeogenic factors in fetuses and reduced in vivo adipose tissue insulin sensitivity in adulthood. In males, undernutrition in early gestation increased adipose tissue lipid handling mechanisms (lipoprotein lipase, glucocorticoid receptor expression) and hepatic methylation within the imprinted control region of insulin-like growth factor 2 receptor in adulthood. Therefore, undernutrition during development induces changes in mechanisms of lipid and glucose metabolism which differ between tissues and sexes dependent on the period of nutritional restriction. Such changes may increase later life obesity and dyslipidaemia risk.
Collapse
Affiliation(s)
- Kirsten R. Poore
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- * E-mail:
| | - Lisa J. Hollis
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Robert J. S. Murray
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Anna Warlow
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Andrew Brewin
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Laurence Fulford
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Jane K. Cleal
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Karen A. Lillycrop
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Graham C. Burdge
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Mark A. Hanson
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Lucy R. Green
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
37
|
Hack M, Schluchter M, Margevicius S, Andreias L, Taylor G, Cuttler L. Trajectory and correlates of growth of extremely-low-birth-weight adolescents. Pediatr Res 2014; 75:358-66. [PMID: 24216539 PMCID: PMC3992255 DOI: 10.1038/pr.2013.209] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 06/19/2013] [Indexed: 12/31/2022]
Abstract
BACKGROUND Catch-up growth may predispose to obesity and metabolic sequelae. We sought to examine the trajectory and correlates of growth and catch up among extremely-low-birth-weight (ELBW) (<1 kg) adolescents. METHODS A cohort study of 148 neurologically normal ELBW children and 115 normal-birth-weight (NBW) controls born during the period 1992-1995 was conducted. Longitudinal measures of gender-specific growth of ELBW children from birth, in addition to growth and measures of obesity of ELBW and NBW children at 14 y, were evaluated. RESULTS Following neonatal growth failure, ELBW children had accelerated growth, but at 8 y, they still had lower weight and height z scores than NBW children. By 14 y, ELBW boys had caught up in growth to their NBW controls, but ELBW girls remained significantly smaller. ELBW children, however, did not differ from their controls in measures of obesity. In hierarchical multiple regression analyses, only maternal BMI and weight gain during infancy and childhood predicted the ELBW children's 14-y weight z scores, BMI z scores, and abdominal circumference. Perinatal risk factors, including intrauterine growth, only predicted growth up to 20 mo. CONCLUSION Maternal BMI and rate of growth, rather than perinatal factors, predict 14-y obesity among neurologically normal ELBW adolescents.
Collapse
Affiliation(s)
- Maureen Hack
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - Mark Schluchter
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, Ohio
| | - Seunghee Margevicius
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, Ohio
| | - Laura Andreias
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - Gerry Taylor
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - Leona Cuttler
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
38
|
O'Reilly M, Hansbro PM, Horvat JC, Beckett EL, Harding R, Sozo F. Bronchiolar remodeling in adult mice following neonatal exposure to hyperoxia: relation to growth. Anat Rec (Hoboken) 2014; 297:758-69. [PMID: 24443274 DOI: 10.1002/ar.22867] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 11/30/2013] [Indexed: 11/07/2022]
Abstract
Preterm infants who receive supplemental oxygen for prolonged periods are at increased risk of impaired lung function later in life. This suggests that neonatal hyperoxia induces persistent changes in small conducting airways (bronchioles). Although the effects of neonatal hyperoxia on alveolarization are well documented, little is known about its effects on developing bronchioles. We hypothesized that neonatal hyperoxia would remodel the bronchiolar walls, contributing to altered lung function in adulthood. We studied three groups of mice (C57BL/6J) to postnatal day 56 (P56; adulthood) when they either underwent lung function testing or necropsy for histological analysis of the bronchiolar wall. One group inhaled 65% O2 from birth until P7, after which they breathed room air; this group experienced growth restriction (HE+GR group). We also used a group in which hyperoxia-induced GR was prevented by dam rotation (HE group). A control group inhaled room air from birth. At P56, the bronchiolar epithelium of HE mice contained fewer Clara cells and more ciliated cells, and the bronchiolar wall contained ∼25% less collagen than controls; in HE+GR mice the bronchiolar walls had ∼13% more collagen than controls. Male HE and HE+GR mice had significantly thicker bronchiolar epithelium than control males and altered lung function (HE males: greater dynamic compliance; HE+GR males: lower dynamic compliance). We conclude that neonatal hyperoxia remodels the bronchiolar wall and, in adult males, affects lung function, but effects are altered by concomitant growth restriction. Our findings may partly explain the reports of poor lung function in ex-preterm children and adults.
Collapse
Affiliation(s)
- Megan O'Reilly
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, 3800, Australia
| | | | | | | | | | | |
Collapse
|
39
|
da Silva Aragão R, Guzmán-Quevedo O, Pérez-García G, Toscano AE, Gois Leandro C, Manhães-de-Castro R, Bolaños-Jiménez F. Differential developmental programming by early protein restriction of rat skeletal muscle according to its fibre-type composition. Acta Physiol (Oxf) 2014; 210:70-83. [PMID: 23362831 DOI: 10.1111/apha.12073] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 11/13/2012] [Accepted: 01/25/2013] [Indexed: 12/29/2022]
Abstract
AIMS Differences in fibre-type composition of skeletal muscle have been associated with obesity and insulin resistance. As a poor nutrient environment early in life is a predisposing factor for the development of obesity and related metabolic diseases at adulthood, this study aimed at determining the long-term consequences of maternal undernutrition on the structural and metabolic properties of two skeletal muscles characterized by their different fibre-type composition and metabolic properties. METHODS The fibre-type composition and enzymatic activities of hexokinase (HK), beta-hydroxyacyl-CoA dehydrogenase (β-HAD) and citrate synthase (CS) were measured in soleus and extensor digitorum longus (EDL) muscles from adult rats born to dams fed a control (17% protein) or a low-protein [8% protein (PR)] diet throughout pregnancy and lactation. In addition, the expression levels of several genes regulating glycolysis, fatty acid oxidation and mitochondrial biogenesis were determined by real-time PCR. RESULTS Protein rats exhibited enhanced density of type II fibres along with decreased rate of fatty acid oxidation and glycolysis in soleus but not EDL. Malnourished rats exhibited also a different gene expression profile in soleus and EDL. Altogether, these alterations correspond to a state of energy deficiency and are present in animals which do not show yet any sign of obesity or glucose intolerance. CONCLUSION We conclude that maternal protein restriction alters in the long term the structural and enzymatic properties of offspring skeletal muscle in a fibre-type-dependent manner. These alterations might have a causative role in the development of obesity and related metabolic disorders later in life.
Collapse
Affiliation(s)
- R. da Silva Aragão
- INRA; UMR1280 Physiologie des Adaptations Nutritionnelles; Nantes France
- Université de Nantes; Nantes Atlantique Université; Nantes France
- Departamento de Nutrição; Centro de Ciências da Saúde; Universidade Federal de Pernambuco; Recife Brazil
| | - O. Guzmán-Quevedo
- INRA; UMR1280 Physiologie des Adaptations Nutritionnelles; Nantes France
- Université de Nantes; Nantes Atlantique Université; Nantes France
| | - G. Pérez-García
- INRA; UMR1280 Physiologie des Adaptations Nutritionnelles; Nantes France
- Université de Nantes; Nantes Atlantique Université; Nantes France
| | - A. E. Toscano
- Núcleo de Enfermagem; CAV; Universidade Federal de Pernambuco; Vitória de Santo Antão Brazil
| | - C. Gois Leandro
- Núcleo de Educação Física e Ciências do Esporte; CAV; Universidade Federal de Pernambuco; Vitória de Santo Antão Brazil
| | - R. Manhães-de-Castro
- Departamento de Nutrição; Centro de Ciências da Saúde; Universidade Federal de Pernambuco; Recife Brazil
| | - F. Bolaños-Jiménez
- INRA; UMR1280 Physiologie des Adaptations Nutritionnelles; Nantes France
- Université de Nantes; Nantes Atlantique Université; Nantes France
| |
Collapse
|
40
|
Brennan KM, Samuel RS, Graugnard TA, Xiao R, Cantor AH, Pescatore AJ. Organic trace mineral levels in the first 96-h post-hatch impact growth performance and intestinal gene expression in broiler chicks. Biol Trace Elem Res 2013; 156:166-74. [PMID: 24078312 DOI: 10.1007/s12011-013-9813-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 09/02/2013] [Indexed: 01/06/2023]
Abstract
Alterations in nutrient intake in the avian neonatal posthatch period can impact development, performance, and metabolism in adulthood. Very little is known about how mineral levels during the post-hatch period affect or “program” gene expression patterns later in life. The objective of this study was to determine the effect of post-hatch (0 to 96 h) dietary mineral supplementation on performance, tissue mineral content, and intestinal gene expression profiles in 21-day-old broiler chicks. One-day-old chicks were randomly assigned to one of two treatment groups consisting of N (organic Zn, Cu, and Mn provided at 100 % of recommendations (National Research Council 1994)) and/or L (organic Zn, Cu, and Mn provided at 20 % of recommendations (National Research Council 1994)) diets fed in two intervals (days 1–4, days 5–21) as follows: (1)N–Lor (2)L–L. Performance parameters did not differ between treatments except that body weight gain was greater (P < 0.05) in L–L birds than N–L birds over the experimental period. Bone mineral content was similar for both treatments at day 21. Intestinal gene expression profiling was examined using the Affymetrix GeneChip Chicken genome array. Ingenuity pathway analysis revealed differences in gene expression profiles between N and L treatments at day 5. At day 21, profiles were unique between N–L and L–L, suggesting that the diet fed until day 4 had an impact on gene expression patterns at day 21 even when birds were fed the same diets day 5–day 21. In this study, we demonstrated that diets fed for the 96 h post-hatch had long-term effects on gene expression, providing unique information as to why post-hatch diets are so important for the longterm bird health and productivity.
Collapse
|
41
|
Lessans S, Dorsey SG. The role for epigenetic modifications in pain and analgesia response. Nurs Res Pract 2013; 2013:961493. [PMID: 24228176 PMCID: PMC3817675 DOI: 10.1155/2013/961493] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 09/12/2013] [Indexed: 12/12/2022] Open
Abstract
Pain remains a poorly understood and managed symptom. A limited mechanistic understanding of interindividual differences in pain and analgesia response shapes current approaches to assessment and treatment. Opportunities exist to improve pain care through increased understanding of how dynamic epigenomic remodeling shapes injury, illness, pain, and treatment response. Tightly regulated alterations of the DNA-histone chromatin complex enable cells to control transcription, replication, gene expression, and protein production. Pathological alterations to chromatin shape the ability of the cell to respond to physiologic and environmental cues leading to disease and reduced treatment effectiveness. This review provides an overview of critical epigenetic processes shaping pathology and pain, highlights current research support for the role of epigenomic modification in the development of chronic pain, and summarizes the therapeutic potential to alter epigenetic processes to improve health outcomes.
Collapse
Affiliation(s)
- Sherrie Lessans
- School of Nursing, University of Maryland, Baltimore, USA
- Program in Neuroscience, University of Maryland, Baltimore, USA
| | - Susan G. Dorsey
- School of Nursing, University of Maryland, Baltimore, USA
- Program in Neuroscience, University of Maryland, Baltimore, USA
| |
Collapse
|
42
|
Ortiz-Espejo M, Pérez-Navero JL, Olza J, Muñoz-Villanueva MC, Aguilera CM, Gil-Campos M. Changes in plasma adipokines in prepubertal children with a history of extrauterine growth restriction. Nutrition 2013; 29:1321-5. [PMID: 24012390 DOI: 10.1016/j.nut.2013.04.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 04/08/2013] [Accepted: 04/19/2013] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Because nutritional support in perinatal life has been associated with metabolic programming, children with a history of extrauterine growth restriction (EUGR) might display alterations in the adipocyte and in the secretion of adipokines. The aim of this study was to assess adiponectin, resistin, and leptin concentrations in prepubertal children with a history of EUGR, and to determine the potential correlation between these adipokines and metabolic parameters. METHODS This case-control study sample included 38 prepubertal children with a history of EUGR and a control group of 123 healthy children of similar age and sex. Anthropometric measures and blood pressure were assessed. Biochemical markers and blood adipokine concentrations (adiponectin, resistin, and leptin) were evaluated. RESULTS Adiponectin concentration was significantly lower in the EUGR group compared with controls (EUGR: 11.49 ± 6.07 versus control: 25.72 ± 10.13 μg/mL), and resistin concentration was higher (EUGR: 20332.95 ± 6401.25 versus control: 8056.31 ± 3823.63 pg/mL), even after adjustment for gestational age, weight, and size at birth. Systolic blood pressure was associated with adipokines concentrations in the EUGR group (P < 0.001). In EUGR children adiponectin was associated with high-density lipoprotein cholesterol (P = 0.042), whereas resistin was associated with carbohydrate metabolism parameters (P < 0.001). CONCLUSIONS Early postnatal malnutrition in EUGR children could program adipose tissue. Plasma adipokines can be measured in childhood to identify precocious changes that may be associated with a higher risk for metabolic syndrome or cardiovascular disease later in life.
Collapse
Affiliation(s)
- María Ortiz-Espejo
- Unit of Metabolism and Pediatric Investigation, Department of Pediatrics, University Reina Sofia Hospital, Córdoba, Spain
| | | | | | | | | | | |
Collapse
|
43
|
Ortiz-Espejo M, Pérez-Navero JL, Muñoz-Villanueva MC, Mercedes GC. Nutritional assessment in neonatal and prepubertal children with a history of extrauterine growth restriction. Early Hum Dev 2013; 89:763-8. [PMID: 23827379 DOI: 10.1016/j.earlhumdev.2013.06.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 05/02/2013] [Accepted: 06/09/2013] [Indexed: 01/22/2023]
Abstract
BACKGROUND Nutritional deficit during perinatal stage may induce significant alterations in adipose tissue and increase the risk of obesity, metabolic syndrome and cardiovascular disease in children with a history of extrauterine growth restriction (EUGR). AIMS To describe the nutritional status in neonatal and prepubertal with a history of EUGR and establish an association between EUGR and later conditions. STUDY DESIGN Descriptive, analytical, observational case-control study. SUBJECTS The study included a sample of 38 prepubertal children with a history of EUGR, and 123 gender-and-age matched controls. OUTCOME MEASURES The EUGR group was asked to answer a food frequency questionnaire. Analysis of body composition in both groups included anthropometric measurements, assessment of blood pressure and biochemical markers. RESULTS Newborns with EUGR received parenteral feeding with a standard nutritional regime and long-chain fatty acid support for 41 ± 23 days; enteral feeding with a special formula for premature infants was initiated at 7 ± 11 days of life. At the prepubertal stage, daily fiber and fatty acid intake in children who had experienced EUGR in the neonatal stage was below the recommended intake. In the EUGR group, the intake of vegetables, fruits and olive oil was below dietary recommendations, while the intake of butchery, fatty meats, pastries and snacks was above the recommendations for the Spanish population. CONCLUSIONS Appropriate nutrition education strategies should be developed for children with a history of EUGR to prevent later associated pathologies, as neonatal nutritional support and feeding during childhood are associated with an increase in diseases in this risk group.
Collapse
Affiliation(s)
- María Ortiz-Espejo
- Unit of Metabolism and Pediatric Investigation, Department of Pediatrics, University Reina Sofia Hospital, Avda Menéndez Pidal s/n., 14004 Córdoba, Spain
| | | | | | | |
Collapse
|
44
|
Ortiz-Espejo M, Gil-Campos M, Mesa MD, García-Rodríguez CE, Muñoz-Villanueva MC, Pérez-Navero JL. Alterations in the antioxidant defense system in prepubertal children with a history of extrauterine growth restriction. Eur J Nutr 2013; 53:607-15. [PMID: 23925485 DOI: 10.1007/s00394-013-0569-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 07/24/2013] [Indexed: 01/24/2023]
Abstract
PURPOSE The role of oxidative stress is well known in the pathogenesis of acquired malnutrition. Intrauterine growth restriction has been associated with an imbalance in oxidative stress/antioxidant system. Therefore, early postnatal environment and, consequently, extrauterine growth restriction might be associated with alterations in the antioxidant defense system, even in the prepubertal stage. METHODS This is a descriptive, analytical, and observational case-control study. The study included two groups; 38 Caucasian prepubertal children born prematurely and with a history of extrauterine growth restriction as the case group, and 123 gender- and age-matched controls. Plasma exogenous antioxidant (retinol, β-carotene, and α-tocopherol) concentrations were measured by HPLC; antioxidant enzyme activities of catalase, glutathione reductase, glutathione peroxidase, and superoxide dismutase were determined in lysed erythrocytes by spectrophotometric techniques. RESULTS Catalase and glutathione peroxidase concentrations were significantly lower in extrauterine growth restriction children than in controls (P < 0.001). Lower plasma retinol concentrations were found in the case group (P = 0.029), while concentrations of β-carotene and α-tocopherol were higher (P < 0.001) in extrauterine growth restriction prepubertal children as compared with controls. After correction by gestational age, birth weight, and length, statistically significant differences were also found, except for retinol. CONCLUSIONS Prepubertal children with a history of extrauterine growth restriction present alterations in their antioxidant defense system. Knowing these alterations may be important in establishing pharmacological and nutritional treatments as this situation might be associated with higher metabolic disorders in adulthood.
Collapse
Affiliation(s)
- M Ortiz-Espejo
- Unit of Metabolism and Pediatric Investigation, Department of Pediatrics, University Reina Sofia Hospital, Avda Menéndez Pidal s/n, 14004, Córdoba, Spain
| | | | | | | | | | | |
Collapse
|
45
|
Bartol FF, Wiley AA, Miller DJ, Silva AJ, Roberts KE, Davolt MLP, Chen JC, Frankshun AL, Camp ME, Rahman KM, Vallet JL, Bagnell CA. LACTATION BIOLOGY SYMPOSIUM: Lactocrine signaling and developmental programming1,2. J Anim Sci 2013; 91:696-705. [DOI: 10.2527/jas.2012-5764] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- F. F. Bartol
- Department of Anatomy, Physiology and Pharmacology, Cellular and Molecular Biosciences Program, Auburn University, Auburn, AL 36849-5517
| | - A. A. Wiley
- Department of Anatomy, Physiology and Pharmacology, Cellular and Molecular Biosciences Program, Auburn University, Auburn, AL 36849-5517
| | - D. J. Miller
- Department of Anatomy, Physiology and Pharmacology, Cellular and Molecular Biosciences Program, Auburn University, Auburn, AL 36849-5517
| | - A. J. Silva
- Department of Anatomy, Physiology and Pharmacology, Cellular and Molecular Biosciences Program, Auburn University, Auburn, AL 36849-5517
| | - K. E. Roberts
- Department of Anatomy, Physiology and Pharmacology, Cellular and Molecular Biosciences Program, Auburn University, Auburn, AL 36849-5517
| | - M. L. P. Davolt
- Department of Anatomy, Physiology and Pharmacology, Cellular and Molecular Biosciences Program, Auburn University, Auburn, AL 36849-5517
| | - J. C. Chen
- Department of Animal Sciences, Endocrinology and Animal Biosciences Program, Rutgers, The State University of New Jersey, New Brunswick 08901
| | - A.-L. Frankshun
- Department of Animal Sciences, Endocrinology and Animal Biosciences Program, Rutgers, The State University of New Jersey, New Brunswick 08901
| | - M. E. Camp
- Department of Animal Sciences, Endocrinology and Animal Biosciences Program, Rutgers, The State University of New Jersey, New Brunswick 08901
| | - K. M. Rahman
- Department of Animal Sciences, Endocrinology and Animal Biosciences Program, Rutgers, The State University of New Jersey, New Brunswick 08901
| | - J. L. Vallet
- USDA ARS, US Meat Animal Research Center, Clay Center, NE 68933-0166
| | - C. A. Bagnell
- Department of Animal Sciences, Endocrinology and Animal Biosciences Program, Rutgers, The State University of New Jersey, New Brunswick 08901
| |
Collapse
|
46
|
|
47
|
Steward DK. Growth Outcomes of Preterm Infants in the Neonatal Intensive Care Unit: Long-term Considerations. ACTA ACUST UNITED AC 2012. [DOI: 10.1053/j.nainr.2012.09.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
48
|
Abstract
AbstractThe science of genetics is undergoing a paradigm shift. Recent discoveries, including the activity of retrotransposons, the extent of copy number variations, somatic and chromosomal mosaicism, and the nature of the epigenome as a regulator of DNA expressivity, are challenging a series of dogmas concerning the nature of the genome and the relationship between genotype and phenotype. According to three widely held dogmas, DNA is the unchanging template of heredity, is identical in all the cells and tissues of the body, and is the sole agent of inheritance. Rather than being an unchanging template, DNA appears subject to a good deal of environmentally induced change. Instead of identical DNA in all the cells of the body, somatic mosaicism appears to be the normal human condition. And DNA can no longer be considered the sole agent of inheritance. We now know that the epigenome, which regulates gene expressivity, can be inherited via the germline. These developments are particularly significant for behavior genetics for at least three reasons: First, epigenetic regulation, DNA variability, and somatic mosaicism appear to be particularly prevalent in the human brain and probably are involved in much of human behavior; second, they have important implications for the validity of heritability and gene association studies, the methodologies that largely define the discipline of behavior genetics; and third, they appear to play a critical role in development during the perinatal period and, in particular, in enabling phenotypic plasticity in offspring. I examine one of the central claims to emerge from the use of heritability studies in the behavioral sciences, the principle of minimal shared maternal effects, in light of the growing awareness that the maternal perinatal environment is a critical venue for the exercise of adaptive phenotypic plasticity. This consideration has important implications for both developmental and evolutionary biology.
Collapse
|
49
|
Abstract
Adverse experiences early in life have the potential to disrupt normal brain development and create stress response channels in preterm infants that are different from those observed in term infants. Animal models show that epigenetic modifications mediate the effects of maternal separation and environmental stress on susceptibility to disease and psychobehavioral problems later in life. Epigenetic research has the potential to lead to the identification of biological markers, gene expression profiles, and profile changes that occur overtime in response to early-life experiences. Combined with knowledge gained through the use of advanced technologies, epigenetic studies have the promise to refine our understanding about how the brain matures and functions from multiple perspectives including the effect of the environment on brain growth and maturation. Such an understanding will pave the way for care practices that will allow the premature brain to develop to its full capacity and will lead to the best possible outcomes. Neonatal epigenetic research is emerging and rapidly advancing. As scientists overcome biological, technical, and cost-related challenges, such research has a great potential in determining key environmental factors that affect the preterm genome, allowing for targeted interventions. The purpose of this article is to explore existing literature related to epigenetic mechanisms that potentially mediate the effects of the environment on preterm infant brain development.
Collapse
|
50
|
Ortiz Espejo M, Gil Campos M, Muñoz Villanueva M, Pérez Navero J. Alteraciones metabólicas en prepúberes con retraso del crecimiento extrauterino. An Pediatr (Barc) 2012; 77:247-53. [DOI: 10.1016/j.anpedi.2012.02.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 01/25/2012] [Accepted: 02/10/2012] [Indexed: 10/28/2022] Open
|