Clinical outcome and gut development after insulin-like growth factor-1 supplementation to preterm pigs

Publikation: Bidrag til tidsskriftTidsskriftartikelForskningfagfællebedømt

Standard

Clinical outcome and gut development after insulin-like growth factor-1 supplementation to preterm pigs. / Holgersen, Kristine; Rasmussen, Martin Bo; Carey, Galen; Burrin, Douglas G.; Thymann, Thomas; Sangild, Per Torp.

I: Frontiers in Pediatrics, Bind 10, 868911, 2022.

Publikation: Bidrag til tidsskriftTidsskriftartikelForskningfagfællebedømt

Harvard

Holgersen, K, Rasmussen, MB, Carey, G, Burrin, DG, Thymann, T & Sangild, PT 2022, 'Clinical outcome and gut development after insulin-like growth factor-1 supplementation to preterm pigs', Frontiers in Pediatrics, bind 10, 868911. https://doi.org/10.3389/fped.2022.868911

APA

Holgersen, K., Rasmussen, M. B., Carey, G., Burrin, D. G., Thymann, T., & Sangild, P. T. (2022). Clinical outcome and gut development after insulin-like growth factor-1 supplementation to preterm pigs. Frontiers in Pediatrics, 10, [868911]. https://doi.org/10.3389/fped.2022.868911

Vancouver

Holgersen K, Rasmussen MB, Carey G, Burrin DG, Thymann T, Sangild PT. Clinical outcome and gut development after insulin-like growth factor-1 supplementation to preterm pigs. Frontiers in Pediatrics. 2022;10. 868911. https://doi.org/10.3389/fped.2022.868911

Author

Holgersen, Kristine ; Rasmussen, Martin Bo ; Carey, Galen ; Burrin, Douglas G. ; Thymann, Thomas ; Sangild, Per Torp. / Clinical outcome and gut development after insulin-like growth factor-1 supplementation to preterm pigs. I: Frontiers in Pediatrics. 2022 ; Bind 10.

Bibtex

@article{ffdeadad8e6c470a97c053f6bbbf703a,
title = "Clinical outcome and gut development after insulin-like growth factor-1 supplementation to preterm pigs",
abstract = "Background: Elevation of circulating insulin-like growth factor-1 (IGF-1) within normal physiological levels may alleviate several morbidities in preterm infants but safety and efficacy remain unclear. We hypothesized that IGF-1 supplementation during the first 1–2 weeks after preterm birth improves clinical outcomes and gut development, using preterm pigs as a model for infants. Methods: Preterm pigs were given vehicle or recombinant human IGF-1/binding protein-3 (rhIGF-1, 2.25 mg/kg/d) by subcutaneous injections for 8 days (Experiment 1, n = 34), or by systemic infusion for 4 days (Experiment 2, n = 19), before collection of blood and organs for analyses. Results: In both experiments, rhIGF-1 treatment increased plasma IGF-1 levels 3-4 fold, reaching the values reported for term suckling piglets. In Experiment 1, rhIGF-1 treatment increased spleen and intestinal weights without affecting clinical outcomes like growth, blood biochemistry (except increased sodium and gamma-glutamyltransferase levels), hematology (e.g., red and white blood cell populations), glucose homeostasis (e.g., basal and glucose-stimulated insulin and glucose levels) or systemic immunity variables (e.g., T cell subsets, neutrophil phagocytosis, LPS stimulation, bacterial translocation to bone marrow). The rhIGF-1 treatment increased gut protein synthesis (+11%, p < 0.05) and reduced the combined incidence of all-cause mortality and severe necrotizing enterocolitis (NEC, p < 0.05), but had limited effects on intestinal morphology, cell proliferation, cell apoptosis, brush-border enzyme activities, permeability and levels of cytokines (IL-1β, IL-6, IL-8). In Experiment 2, rhIGF-1 treated pigs had reduced blood creatine kinase, creatinine, potassium and aspartate aminotransferase levels, with no effects on organ weights (except increased spleen weight), blood chemistry values, clinical variables or NEC. Conclusion: Physiological elevation of systemic IGF-1 levels for 8 days after preterm birth increased intestinal weight and protein synthesis, spleen weight and potential overall viability of pigs, without any apparent negative effects on recorded clinical parameters. The results add further preclinical support for safety and efficacy of supplemental IGF-1 to hospitalized very preterm infants.",
keywords = "development, gut, insulin-like growth factor-1, pig, preterm",
author = "Kristine Holgersen and Rasmussen, {Martin Bo} and Galen Carey and Burrin, {Douglas G.} and Thomas Thymann and Sangild, {Per Torp}",
note = "Publisher Copyright: Copyright {\textcopyright} 2022 Holgersen, Rasmussen, Carey, Burrin, Thymann and Sangild.",
year = "2022",
doi = "10.3389/fped.2022.868911",
language = "English",
volume = "10",
journal = "Frontiers in Pediatrics",
issn = "2296-2360",
publisher = "Frontiers Media",

}

RIS

TY - JOUR

T1 - Clinical outcome and gut development after insulin-like growth factor-1 supplementation to preterm pigs

AU - Holgersen, Kristine

AU - Rasmussen, Martin Bo

AU - Carey, Galen

AU - Burrin, Douglas G.

AU - Thymann, Thomas

AU - Sangild, Per Torp

N1 - Publisher Copyright: Copyright © 2022 Holgersen, Rasmussen, Carey, Burrin, Thymann and Sangild.

PY - 2022

Y1 - 2022

N2 - Background: Elevation of circulating insulin-like growth factor-1 (IGF-1) within normal physiological levels may alleviate several morbidities in preterm infants but safety and efficacy remain unclear. We hypothesized that IGF-1 supplementation during the first 1–2 weeks after preterm birth improves clinical outcomes and gut development, using preterm pigs as a model for infants. Methods: Preterm pigs were given vehicle or recombinant human IGF-1/binding protein-3 (rhIGF-1, 2.25 mg/kg/d) by subcutaneous injections for 8 days (Experiment 1, n = 34), or by systemic infusion for 4 days (Experiment 2, n = 19), before collection of blood and organs for analyses. Results: In both experiments, rhIGF-1 treatment increased plasma IGF-1 levels 3-4 fold, reaching the values reported for term suckling piglets. In Experiment 1, rhIGF-1 treatment increased spleen and intestinal weights without affecting clinical outcomes like growth, blood biochemistry (except increased sodium and gamma-glutamyltransferase levels), hematology (e.g., red and white blood cell populations), glucose homeostasis (e.g., basal and glucose-stimulated insulin and glucose levels) or systemic immunity variables (e.g., T cell subsets, neutrophil phagocytosis, LPS stimulation, bacterial translocation to bone marrow). The rhIGF-1 treatment increased gut protein synthesis (+11%, p < 0.05) and reduced the combined incidence of all-cause mortality and severe necrotizing enterocolitis (NEC, p < 0.05), but had limited effects on intestinal morphology, cell proliferation, cell apoptosis, brush-border enzyme activities, permeability and levels of cytokines (IL-1β, IL-6, IL-8). In Experiment 2, rhIGF-1 treated pigs had reduced blood creatine kinase, creatinine, potassium and aspartate aminotransferase levels, with no effects on organ weights (except increased spleen weight), blood chemistry values, clinical variables or NEC. Conclusion: Physiological elevation of systemic IGF-1 levels for 8 days after preterm birth increased intestinal weight and protein synthesis, spleen weight and potential overall viability of pigs, without any apparent negative effects on recorded clinical parameters. The results add further preclinical support for safety and efficacy of supplemental IGF-1 to hospitalized very preterm infants.

AB - Background: Elevation of circulating insulin-like growth factor-1 (IGF-1) within normal physiological levels may alleviate several morbidities in preterm infants but safety and efficacy remain unclear. We hypothesized that IGF-1 supplementation during the first 1–2 weeks after preterm birth improves clinical outcomes and gut development, using preterm pigs as a model for infants. Methods: Preterm pigs were given vehicle or recombinant human IGF-1/binding protein-3 (rhIGF-1, 2.25 mg/kg/d) by subcutaneous injections for 8 days (Experiment 1, n = 34), or by systemic infusion for 4 days (Experiment 2, n = 19), before collection of blood and organs for analyses. Results: In both experiments, rhIGF-1 treatment increased plasma IGF-1 levels 3-4 fold, reaching the values reported for term suckling piglets. In Experiment 1, rhIGF-1 treatment increased spleen and intestinal weights without affecting clinical outcomes like growth, blood biochemistry (except increased sodium and gamma-glutamyltransferase levels), hematology (e.g., red and white blood cell populations), glucose homeostasis (e.g., basal and glucose-stimulated insulin and glucose levels) or systemic immunity variables (e.g., T cell subsets, neutrophil phagocytosis, LPS stimulation, bacterial translocation to bone marrow). The rhIGF-1 treatment increased gut protein synthesis (+11%, p < 0.05) and reduced the combined incidence of all-cause mortality and severe necrotizing enterocolitis (NEC, p < 0.05), but had limited effects on intestinal morphology, cell proliferation, cell apoptosis, brush-border enzyme activities, permeability and levels of cytokines (IL-1β, IL-6, IL-8). In Experiment 2, rhIGF-1 treated pigs had reduced blood creatine kinase, creatinine, potassium and aspartate aminotransferase levels, with no effects on organ weights (except increased spleen weight), blood chemistry values, clinical variables or NEC. Conclusion: Physiological elevation of systemic IGF-1 levels for 8 days after preterm birth increased intestinal weight and protein synthesis, spleen weight and potential overall viability of pigs, without any apparent negative effects on recorded clinical parameters. The results add further preclinical support for safety and efficacy of supplemental IGF-1 to hospitalized very preterm infants.

KW - development

KW - gut

KW - insulin-like growth factor-1

KW - pig

KW - preterm

U2 - 10.3389/fped.2022.868911

DO - 10.3389/fped.2022.868911

M3 - Journal article

C2 - 35989990

AN - SCOPUS:85136499077

VL - 10

JO - Frontiers in Pediatrics

JF - Frontiers in Pediatrics

SN - 2296-2360

M1 - 868911

ER -

ID: 319164250