Postnatal serum IGF-1 levels associate with brain volumes at term in extremely preterm infants

William Hellström, Lisa M Hortensius, Chatarina Löfqvist, Gunnel Hellgren, Maria Luisa Tataranno, David Ley, Manon J N L Benders, Ann Hellström, Isabella M Björkman-Burtscher, Rolf A Heckemann, Karin Sävman, William Hellström, Lisa M Hortensius, Chatarina Löfqvist, Gunnel Hellgren, Maria Luisa Tataranno, David Ley, Manon J N L Benders, Ann Hellström, Isabella M Björkman-Burtscher, Rolf A Heckemann, Karin Sävman

Abstract

Background: Growth factors important for normal brain development are low in preterm infants. This study investigated the link between growth factors and preterm brain volumes at term.

Material/methods: Infants born <28 weeks gestational age (GA) were included. Endogenous levels of insulin-like growth factor (IGF)-1, brain-derived growth factor, vascular endothelial growth factor, and platelet-derived growth factor (expressed as area under the curve [AUC] for serum samples from postnatal days 1, 7, 14, and 28) were utilized in a multivariable linear regression model. Brain volumes were determined by magnetic resonance imaging (MRI) at term equivalent age.

Results: In total, 49 infants (median [range] GA 25.4 [22.9-27.9] weeks) were included following MRI segmentation quality assessment and AUC calculation. IGF-1 levels were independently positively associated with the total brain (p < 0.001, β = 0.90), white matter (p = 0.007, β = 0.33), cortical gray matter (p = 0.002, β = 0.43), deep gray matter (p = 0.008, β = 0.05), and cerebellar (p = 0.006, β = 0.08) volume adjusted for GA at birth and postmenstrual age at MRI. No associations were seen for other growth factors.

Conclusions: Endogenous exposure to IGF-1 during the first 4 weeks of life was associated with total and regional brain volumes at term. Optimizing levels of IGF-1 might improve brain growth in extremely preterm infants.

Impact: High serum levels of insulin-like growth factor (IGF)-1 during the first month of life were independently associated with increased total brain volume, white matter, gray matter, and cerebellar volume at term equivalent age in extremely preterm infants. IGF-1 is a critical regulator of neurodevelopment and postnatal levels are low in preterm infants. The effects of IGF-1 levels on brain development in extremely preterm infants are not fully understood. Optimizing levels of IGF-1 may benefit early brain growth in extremely preterm infants. The effects of systemically administered IGF-1/IGFBP3 in extremely preterm infants are now being investigated in a randomized controlled trial (Clinicaltrials.gov: NCT03253263).

Conflict of interest statement

A.H. and C.L. hold stock/stock options in Premalux AB. In addition, A.H. and D.L. have received consulting fees from Shire, a Takeda company. The rest of the authors declare no competing interests.

© 2022. The Author(s).

Figures

Fig. 1. Consort study flowchart.
Fig. 1. Consort study flowchart.
In total, 49 infants were eligible for analysis with complete MRI volume segmentation and growth factor AUC. *In two infants, IGF-1 AUC was available, but not BDNF, VEGF, and PDGF. RCT randomized controlled trial, n number, TEA term equivalent age, IGF Insulin-like growth factor, BDNF brain-derived neurotrophic factor, AUC area under the curve, MRI magnetic resonance imaging.
Fig. 2. Serum levels of IGF-1 AUC…
Fig. 2. Serum levels of IGF-1 AUC (PND 1–28) and regional brain volumes at TEA.
Higher IGF-1 levels were associated with total brain volume; r = 0.49, p < 0.001 (a), white matter volume; r = 0.55, p < 0.001 (b), cortical gray matter volume; r = 0.40, p = 0.004 (c), deep gray matter volume r = 0.45, p = 0.001 (d), and cerebellar volume; r = 0.47, p < 0.001 (e). IGF insulin-like growth factor, AUC area under the curve, PND postnatal day, TEA term equivalent age.
Fig. 3. The impact of gestational age…
Fig. 3. The impact of gestational age at birth on brain volume and serum IGF-1 levels.
a Estimated unstandardized probabilities of total brain volume, retrieved from the full statistical model, illustrated per gestational week. Error bars indicate 95% CI. Colored area: interpolated. b Distribution of serum levels of IGF-1 during postnatal days 1, 7, 14, and 28 in relation to gestational age at birth (below or over 25 weeks). More immature infants show less increase in endogenous IGF-1 levels. Dotted line shows 30 ng/ml. A prolonged period with serum IGF-1 levels below this threshold has been related to morbidities in the neurovascular unit. Boxes illustrate interquartile range, whiskers show full range. IGF-1 insulin-like growth factor 1, CI confidence interval, PND postnatal day, GA gestational age.

References

    1. Ball G, et al. Development of cortical microstructure in the preterm human brain. Proc. Natl Acad. Sci. USA. 2013;110:9541–9546. doi: 10.1073/pnas.1301652110.
    1. Smyser CD, et al. Resting-state network complexity and magnitude are reduced in prematurely born infants. Cereb. Cortex. 2016;26:322–333. doi: 10.1093/cercor/bhu251.
    1. Scheinost D, et al. Preterm birth alters neonatal, functional rich club organization. Brain Struct. Funct. 2016;221:3211–3222. doi: 10.1007/s00429-015-1096-6.
    1. Guihard-Costa AM, Larroche JC. Differential growth between the fetal brain and its infratentorial part. Early Hum. Dev. 1990;23:27–40. doi: 10.1016/0378-3782(90)90126-4.
    1. Lind A, et al. Associations between regional brain volumes at term-equivalent age and development at 2 years of age in preterm children. Pediatr. Radiol. 2011;41:953–961. doi: 10.1007/s00247-011-2071-x.
    1. Thompson DK, et al. Characterisation of brain volume and microstructure at term-equivalent age in infants born across the gestational age spectrum. NeuroImage Clin. 2019;21:101630. doi: 10.1016/j.nicl.2018.101630.
    1. Storbeck T, Bruns N, Weiss C, Felderhoff-Müser U, Müller H. Correlation of lateral ventricular size and deep gray matter volume in MRI at term equivalent age with neurodevelopmental outcome at a corrected age of 24 months and with handedness in preterm infants. Eur. J. Pediatr. 2020;179:271–278. doi: 10.1007/s00431-019-03496-4.
    1. Peterson BS, et al. Regional brain volume abnormalities and long-term cognitive outcome in preterm infants. JAMA. 2000;284:1939–1947. doi: 10.1001/jama.284.15.1939.
    1. Kelly CE, et al. Regional brain volumes, microstructure and neurodevelopment in moderate–late preterm children. Arch. Dis. Child. Fetal Neonatal Ed. 2020;105:593. doi: 10.1136/archdischild-2019-317941.
    1. Hellstrom A, et al. Insulin-like growth factor 1 has multisystem effects on foetal and preterm infant development. Acta Paediatr. 2016;105:576–586. doi: 10.1111/apa.13350.
    1. Hansen-Pupp I, et al. Circulatory insulin-like growth factor-I and brain volumes in relation to neurodevelopmental outcome in very preterm infants. Pediatr. Res. 2013;74:564–569. doi: 10.1038/pr.2013.135.
    1. Hansen-Pupp I, et al. Postnatal decrease in circulating insulin-like growth factor-I and low brain volumes in very preterm infants. J. Clin. Endocrinol. Metab. 2011;96:1129–1135. doi: 10.1210/jc.2010-2440.
    1. Cai Z, Fan LW, Lin S, Pang Y, Rhodes PG. Intranasal administration of insulin-like growth factor-1 protects against lipopolysaccharide-induced injury in the developing rat brain. Neuroscience. 2011;194:195–207. doi: 10.1016/j.neuroscience.2011.08.003.
    1. Lin S, et al. IGF-1 protects oligodendrocyte progenitor cells and improves neurological functions following cerebral hypoxia-ischemia in the neonatal rat. Brain Res. 2005;1063:15–26. doi: 10.1016/j.brainres.2005.09.042.
    1. Brywe KG, et al. IGF-I neuroprotection in the immature brain after hypoxia-ischemia, involvement of Akt and GSK3beta? Eur. J. Neurosci. 2005;21:1489–1502. doi: 10.1111/j.1460-9568.2005.03982.x.
    1. Cao Y, et al. Insulin-like growth factor (IGF)-1 suppresses oligodendrocyte caspase-3 activation and increases glial proliferation after ischemia in near-term fetal sheep. J. Cereb. Blood Flow. Metab. 2003;23:739–747. doi: 10.1097/01.WCB.0000067720.12805.6F.
    1. Miranda M., Morici J. F., Zanoni M. B. & Bekinschtein P. Brain-derived neurotrophic factor: a key molecule for memory in the healthy and the pathological brain. Front. Cell. Neurosci. 13, 363 (2019).
    1. Ogunshola OO, et al. Neuronal VEGF expression correlates with angiogenesis in postnatal developing rat brain. Brain Res. Dev. Brain Res. 2000;119:139–153. doi: 10.1016/S0165-3806(99)00125-X.
    1. Feng Y, Rhodes PG, Bhatt AJ. Neuroprotective effects of vascular endothelial growth factor following hypoxic ischemic brain injury in neonatal rats. Pediatr. Res. 2008;64:370–374. doi: 10.1203/PDR.0b013e318180ebe6.
    1. Funa K, Sasahara M. The roles of PDGF in development and during neurogenesis in the normal and diseased nervous system. J. Neuroimmune Pharmacol. 2014;9:168–181. doi: 10.1007/s11481-013-9479-z.
    1. . Fatty Acids Study in Preventing Retinopathy of Prematurity. NCT02760472. . Accessed November 2019.
    1. Najm S, et al. Effects of a lipid emulsion containing fish oil on polyunsaturated fatty acid profiles, growth and morbidities in extremely premature infants: a randomized controlled trial. Clin. Nutr. Espen. 2017;20:17–23. doi: 10.1016/j.clnesp.2017.04.004.
    1. Blum WF, Breier BH. Radioimmunoassays for IGFs and IGFBPs. Growth Regul. 1994;4(Suppl 1):11–19.
    1. Löfqvist C, et al. A pharmacokinetic and dosing study of intravenous insulin-like growth factor-I and IGF-binding protein-3 complex to preterm infants. Pediatr. Res. 2009;65:574–579. doi: 10.1203/PDR.0b013e31819d9e8c.
    1. Hellgren G, et al. Decreased platelet counts and serum levels of VEGF-A, PDGF-BB, and BDNF in extremely preterm infants developing severe ROP. Neonatology. 2021;118:18–27. doi: 10.1159/000512282.
    1. Makropoulos A, et al. The developing human connectome project: a minimal processing pipeline for neonatal cortical surface reconstruction. NeuroImage. 2018;173:88–112. doi: 10.1016/j.neuroimage.2018.01.054.
    1. Gousias IS, et al. Magnetic resonance imaging of the newborn brain: automatic segmentation of brain images into 50 anatomical regions. PLoS One. 2013;8:e59990. doi: 10.1371/journal.pone.0059990.
    1. Makropoulos A, et al. Automatic whole brain MRI segmentation of the developing neonatal brain. IEEE Trans. Med. Imaging. 2014;33:1818–1831. doi: 10.1109/TMI.2014.2322280.
    1. Holm S. A simple sequentially rejective multiple test procedure. Scand. J. Stat. 1979;6:65–70.
    1. Papile LA, Burstein J, Burstein R, Koffler H. Incidence and evolution of subependymal and intraventricular hemorrhage: a study of infants with birth weights less than 1,500 gm. J. Pediatr. 1978;92:529–534. doi: 10.1016/S0022-3476(78)80282-0.
    1. Kidokoro H, Neil JJ, Inder TE. New MR imaging assessment tool to define brain abnormalities in very preterm infants at term. AJNR Am. J. Neuroradiol. 2013;34:2208–2214. doi: 10.3174/ajnr.A3521.
    1. Bach MA, Shen-Orr Z, Lowe WL, Roberts CT, Leroith D. Insulin-like growth factor I mRNA levels are developmentally regulated in specific regions of the rat brain. Mol. Brain Res. 1991;10:43–48. doi: 10.1016/0169-328X(91)90054-2.
    1. Bartlett WP, Li XS, Williams M, Benkovic S. Localization of insulin-like growth factor-1 mRNA in murine central nervous system during postnatal development. Dev. Biol. 1991;147:239–250. doi: 10.1016/S0012-1606(05)80021-1.
    1. Aburto MR, Magariños M, Leon Y, Varela-Nieto I, Sanchez-Calderon H. AKT signaling mediates IGF-I survival actions on otic neural progenitors. PLoS One. 2012;7:e30790. doi: 10.1371/journal.pone.0030790.
    1. Arsenijevic Y, Weiss S, Schneider B, Aebischer P. Insulin-like growth factor-I is necessary for neural stem cell proliferation and demonstrates distinct actions of epidermal growth factor and fibroblast growth factor-2. J. Neurosci. 2001;21:7194–7202. doi: 10.1523/JNEUROSCI.21-18-07194.2001.
    1. Ye P, et al. Astrocyte-specific overexpression of insulin-like growth factor-I promotes brain overgrowth and glial fibrillary acidic protein expression. J. Neurosci. Res. 2004;78:472–484. doi: 10.1002/jnr.20288.
    1. Han VK, D’Ercole AJ, Lund PK. Cellular localization of somatomedin (insulin-like growth factor) messenger RNA in the human fetus. Science. 1987;236:193–197. doi: 10.1126/science.3563497.
    1. Carro E, Nuñez A, Busiguina S, Torres-Aleman I. Circulating insulin-like growth factor I mediates effects of exercise on the brain. J. Neurosci. 2000;20:2926–2933. doi: 10.1523/JNEUROSCI.20-08-02926.2000.
    1. Santi A, Genis L, Torres Aleman I. A coordinated action of blood-borne and brain insulin-like growth factor I in the response to traumatic brain injury. Cereb. Cortex. 2018;28:2007–2014. doi: 10.1093/cercor/bhx106.
    1. Carson MJ, Behringer RR, Brinster RL, McMorris FA. Insulin-like growth factor I increases brain growth and central nervous system myelination in transgenic mice. Neuron. 1993;10:729–740. doi: 10.1016/0896-6273(93)90173-O.
    1. Ye P, Carson J, D’Ercole AJ. In vivo actions of insulin-like growth factor-I (IGF-I) on brain myelination: studies of IGF-I and IGF binding protein-1 (IGFBP-1) transgenic mice. J. Neurosci. 1995;15:7344–7356. doi: 10.1523/JNEUROSCI.15-11-07344.1995.
    1. Sveinsdóttir K, et al. Impaired cerebellar maturation, growth restriction, and circulating insulin-like growth factor 1 in preterm rabbit pups. Dev. Neurosci. 2017;39:487–497. doi: 10.1159/000480428.
    1. Woodward LJ, Anderson PJ, Austin NC, Howard K, Inder TE. Neonatal MRI to predict neurodevelopmental outcomes in preterm infants. N. Engl. J. Med. 2006;355:685–694. doi: 10.1056/NEJMoa053792.
    1. Volpe JJ. Cerebellum of the premature infant: rapidly developing, vulnerable, clinically important. J. Child Neurol. 2009;24:1085–1104. doi: 10.1177/0883073809338067.
    1. Iwata S, et al. Qualitative brain MRI at term and cognitive outcomes at 9 years after very preterm birth. Pediatrics. 2012;129:e1138–e1147. doi: 10.1542/peds.2011-1735.
    1. Spittle AJ, et al. Neonatal white matter abnormality predicts childhood motor impairment in very preterm children. Dev. Med. Child Neurol. 2011;53:1000–1006. doi: 10.1111/j.1469-8749.2011.04095.x.
    1. Katušić A, Raguž M, Žunić Išasegi I. Brain tissue volumes at term-equivalent age are associated with early motor behavior in very preterm infants. Int. J. Dev. Neurosci. 2020;80:409–417. doi: 10.1002/jdn.10039.
    1. Shah DK, et al. Reduction in cerebellar volumes in preterm infants: relationship to white matter injury and neurodevelopment at two years of age. Pediatr. Res. 2006;60:97–102. doi: 10.1203/01.pdr.0000220324.27597.f0.
    1. Spittle AJ, et al. Reduced cerebellar diameter in very preterm infants with abnormal general movements. Early Hum. Dev. 2010;86:1–5. doi: 10.1016/j.earlhumdev.2009.11.002.
    1. Van Kooij BJ, et al. Cerebellar volume and proton magnetic resonance spectroscopy at term, and neurodevelopment at 2 years of age in preterm infants. Dev. Med Child Neurol. 2012;54:260–266. doi: 10.1111/j.1469-8749.2011.04168.x.
    1. Anderson P, Doyle LW. Neurobehavioral outcomes of school-age children born extremely low birth weight or very preterm in the 1990s. JAMA. 2003;289:3264–3272. doi: 10.1001/jama.289.24.3264.
    1. Keunen K, et al. Brain tissue volumes in preterm infants: prematurity, perinatal risk factors and neurodevelopmental outcome: a systematic review. J. Matern. Fetal Neonatal Med. 2012;25:89–100. doi: 10.3109/14767058.2012.664343.
    1. Yumani, D. F. J., Calor A. K. & van Weissenbruch M. M. The course Of IGF-1 levels and nutrient intake in extremely and very preterm infants during hospitalisation. Nutrients12, 675 (2020).
    1. Hellstrom A, et al. Low IGF-I suppresses VEGF-survival signaling in retinal endothelial cells: direct correlation with clinical retinopathy of prematurity. Proc. Natl Acad. Sci. USA. 2001;98:5804. doi: 10.1073/pnas.101113998.
    1. Smith LE, et al. Regulation of vascular endothelial growth factor-dependent retinal neovascularization by insulin-like growth factor-1 receptor. Nat. Med. 1999;5:1390–1395. doi: 10.1038/70963.
    1. Hellström A, et al. IGF-I is critical for normal vascularization of the human retina. J. Clin. Endocrinol. Metab. 2002;87:3413–3416. doi: 10.1210/jcem.87.7.8629.
    1. Hellstrom A, et al. Low IGF-I suppresses VEGF-survival signaling in retinal endothelial cells: direct correlation with clinical retinopathy of prematurity. Proc. Natl Acad. Sci. USA. 2001;98:5804–5808. doi: 10.1073/pnas.101113998.
    1. Begliuomini S, et al. Plasma brain-derived neurotrophic factor daily variations in men: correlation with cortisol circadian rhythm. J. Endocrinol. 2008;197:429–435. doi: 10.1677/JOE-07-0376.
    1. Chouthai NS, Sampers J, Desai N, Smith GM. Changes in neurotrophin levels in umbilical cord blood from infants with different gestational ages and clinical conditions. Pediatr. Res. 2003;53:965–969. doi: 10.1203/01.PDR.0000061588.39652.26.
    1. Kowiański P, et al. BDNF: a key factor with multipotent impact on brain signaling and synaptic plasticity. Cell. Mol. Neurobiol. 2018;38:579–593. doi: 10.1007/s10571-017-0510-4.
    1. Skogstrand K, et al. Reduced neonatal brain-derived neurotrophic factor is associated with autism spectrum disorders. Transl. Psychiatry. 2019;9:252. doi: 10.1038/s41398-019-0587-2.
    1. Ghassabian A, et al. Determinants of neonatal brain-derived neurotrophic factor and association with child development. Dev. Psychopathol. 2017;29:1499–1511. doi: 10.1017/S0954579417000414.
    1. Oak P, Hilgendorff A. The BPD trio? Interaction of dysregulated PDGF, VEGF, and TGF signaling in neonatal chronic lung disease. Mol. Cell. Pediatr. 2017;4:11. doi: 10.1186/s40348-017-0076-8.
    1. Oak P, et al. Attenuated PDGF signaling drives alveolar and microvascular defects in neonatal chronic lung disease. EMBO Mol. Med. 2017;9:1504–1520. doi: 10.15252/emmm.201607308.
    1. Sveinsdóttir K, et al. Relation of retinopathy of prematurity to brain volumes at term equivalent age and developmental outcome at 2 years of corrected age in very preterm infants. Neonatology. 2018;114:46–52. doi: 10.1159/000487847.
    1. Niklasson A, Ericson A, Fryer JG, Karlberg J, Lawrence C, Karlberg P, et al. An update of the Swedish reference standards for weight, length and head circumference at birth for given gestational age (1977-1981) Acta Paediatr Scand. 1991;80:756–62. doi: 10.1111/j.1651-2227.1991.tb11945.x.
    1. International Committee for the Classification of Retinopathy of Prematurity. The International Classification of Retinopathy of Prematurity revisited. Arch Ophthalmol. 123, 991–9 (2005).

Source: PubMed

3
Suscribir