257
Views
0
CrossRef citations to date
0
Altmetric
Review

Early Repetitive Pain in Preterm Infants in Relation to the Developing Brain

&
Pages 57-67 | Published online: 19 Dec 2013

References

  • Blencowe H , CousensS, OestergaardMZ et al. National, regional, and worldwide estimates of preterm birth rates in the year 2010 with time trends since 1990 for selected countries: a systematic analysis and implications. Lancet 379(9832) , 2162–2172 (2009).
  • Martin J , HamiltonB, VenturaS, OstermanM, and Mathiews T. Births final data for 2011. Natl Vital Stat. Rep.62(1) , (2013).
  • Aarnoudse-Moens C , Weisglas-KuperusN, van Goudoever J, Oosterlaan J. Meta-analysis of neurobehavioral outcomes in very preterm and/or low birth weight children. Pediatrics124(2) , 717–728 (2009).
  • Synnes AR , AnsonS, ArkesteijnA et al. School entry age outcomes for infants with birth weight </= 800 grams. J. Pediatr. 157(6) , 989–994 (2010).
  • Grunau RE , WhitfieldMF, Petrie-ThomasJ et al. Neonatal pain, parenting stress and interaction, in relation to cognitive and motor development at 8 and 18 months in preterm infants. Pain 143(1–2) , 138–146 (2009).
  • Volpe JJ . The encephalopathy of prematurity – brain injury and impaired brain development inextricably intertwined. Semin. Pediatr. Neurol.16(4) , 167–178 (2009).
  • Carbajal R , RoussetA, DananC et al. Epidemiology and treatment of painful procedures in neonates in intensive care units. JAMA 300(1) , 60–70 (2008).
  • Simons SH , vanDM, AnandKS, RoofthooftD, van Lingen RA, Tibboel D. Do we still hurt newborn babies? A prospective study of procedural pain and analgesia in neonates. Arch. Pediatr. Adolesc. Med.157(11) , 1058–1064 (2003).
  • Johnston C , BarringtonKJ, TaddioA, CarbajalR, FilionF. Pain in Canadian NICUs: have we improved over the past 12 years? Clin. J. Pain.27(3) , 225–232 (2011).
  • Grunau RE , HolstiL, PetersJWB. Long-term consequences of pain in human neonates. Semin. Fetal. Neonatal. Med.11(4) , 268–275 (2006).
  • Volpe JJ . Brain injury in premature infants: a complex amalgam of destructive and developmental disturbances. Lancet Neurol.8(1) , 110–124 (2009).
  • Anand KJS , ScalzoFM. Can adverse neonatal experiences alter brain development and subsequent behavior? Neonatology77(2) , 69–82 (2000).
  • Grunau R . Early Pain in Preterm Infants: a model of long-term effects. Clin. Perinatol.29 , 373–394 (2002).
  • Brummelte S , GrunauRE, ChauV et al. Procedural pain and brain development in premature newborns. Ann. Neurol. 71(3) , 385–396 (2012).
  • Ranger M , ChauCM, GargA et al. Neonatal pain-related stress predicts cortical thickness at age 7 years in children born very preterm. PLoS ONE 8(10) , e76702 (2013).
  • Kostovic I , Jovanov-MilosevicN. The development of cerebral connections during the first 20–45 weeks‘ gestation. Semin. Fetal. Neonatal. Med.11(6) , 415–422 (2006).
  • Power JD , FairDA, SchlaggarBL, PetersenSE. The development of human functional brain networks. Neuron67(5) , 735–748 (2010).
  • Jackman A , FitzgeraldM. Development of peripheral hindlimb and central spinal cord innervation by subpopulations of dorsal root ganglion cells in the embryonic rat. J. Comp. Neurol.418(3) , 281–298 (2000).
  • Fitzgerald M . The development of nociceptive circuits. Nat. Rev. Neurosci.6(7) , 507–520 (2005).
  • Beggs S , TorsneyC, DrewLJ, FitzgeraldM. The postnatal reorganization of primary afferent input and dorsal horn cell receptive fields in the rat spinal cord is an activity-dependent process. Eur. J. Neurosci.16(7) , 1249–1258 (2002).
  • Fitzgerald M , WalkerSM. Infant pain management: a developmental neurobiological approach. Nat. Clin. Pract. Neuro.5(1) , 35–50 (2009).
  • Knaepen L , PatijnJ, van Kleef M, Mulder M, Tibboel D, Joosten EAJ. Neonatal repetitive needle pricking: Plasticity of the spinal nociceptive circuit and extended postoperative pain in later life. Dev. Neurobiol.73(1) , 85–97 (2013).
  • Anand KJ , CoskunV, ThrivikramanKV, NemeroffCB, PlotskyPM. Long-term behavioral effects of repetitive pain in neonatal rat pups. Physiol. Behav.66(4) , 627–637 (1999).
  • Ruda MA , LingQD, HohmannAG, PengYB, TachibanaT. Altered nociceptive neuronal circuits after neonatal peripheral inflammation. Science289(5479) , 628–630 (2000).
  • Sternberg WF , ScorrL, SmithLD, RidgwayCG, StoutM. Long-term effects of neonatal surgery on adulthood pain behavior. Pain113(3) , 347–353 (2005).
  • Hohmeister J , KrollA, Wollgarten-HadamekI et al. Cerebral processing of pain in school-aged children with neonatal nociceptive input: an exploratory fMRI study. Pain 150(2) , 257–267 (2010).
  • Abdulkader HM , FreerY, GarryEM, Fleetwood-WalkerSM, McIntoshN. Prematurity and neonatal noxious events exert lasting effects on infant pain behaviour. Early Hum. Devel.84(6) , 351–355 (2008).
  • Walker SM , FranckLS, FitzgeraldM, MylesJ, StocksJ, MarlowN. Long-term impact of neonatal intensive care and surgery on somatosensory perception in children born extremely preterm. Pain141(1–2) , 79–87 (2009).
  • Hermann C , HohmeisterJ, DemirakçaS, ZohselK, FlorH. Long-term alteration of pain sensitivity in school-aged children with early pain experiences. Pain125(3) , 278–285 (2006).
  • Johnston CC , StevensBJ. Experience in a neonatal intensive care unit affects pain response. Pediatrics98(5) , 925–930 (1996).
  • Grunau RE , OberlanderTF, WhitfieldMF, FitzgeraldC, LeeSK. Demographic and therapeutic determinants of pain reactivity in very low birth weight neonates at 32 weeks‘ postconceptional Age. Pediatrics107(1) , 105–112 (2001).
  • Grunau RE , HolstiL, HaleyDW et al. Neonatal procedural pain exposure predicts lower cortisol and behavioral reactivity in preterm infants in the NICU. Pain 113(3) , 293–300 (2005).
  • Grunau RV , WhitfieldMF, PetrieJH. Pain sensitivity and temperament in extremely low-birth-weight premature toddlers and preterm and full-term controls. Pain58(3) , 341–346 (1994).
  • Allegaert K , DevliegerH, BulckaertD, NaulaersG, CasaerP, TibboelD. Variability in pain expression characteristics in former preterm infants. J. Perinat. Med.33(5) , 442–448 (2005).
  • Pryce CR , Rüedi-BettschenD, DettlingAC, FeldonJ. Early life stress: long-term physiological impact in rodents and primates. Physiology17(4) , 150–155 (2002).
  • Grunau RV , WhitfieldMF, PetrieJH, FryerEL. Early pain experience, child and family factors, as precursors of somatization: a prospective study of extremely premature and fullterm children. Pain56(3) , 353–359 (1994).
  • Hohmeister J , DemirakcaS, ZohselK, FlorH, HermannC. Responses to pain in school-aged children with experience in a neonatal intensive care unit: Cognitive aspects and maternal influences. Eur. J. Pain.13(1) , 94–101 (2009).
  • Rogers CE , AndersonPJ, ThompsonDK et al. Regional cerebral development at term relates to school-age social-emotional development in very preterm children. J. Am. Acad. Child Adolesc. Psychiatry 51(2) , 181–191 (2012).
  • Miller SP , VigneronDB, HenryRG et al. Serial quantitative diffusion tensor MRI of the premature brain: development in newborns with and without injury. J. Magn. Reson. Imaging 16(6) , 621–632 (2002).
  • Smith GC , GutovichJ, SmyserC et al. Neonatal intensive care unit stress is associated with brain development in preterm infants. Ann. Neurol. 70(4) , 541–549 (2011).
  • Peterson BS , VohrB, StaibLH et al. Regional brain volume abnormalities and long-term cognitive outcome in preterm infants. JAMA 284(15) , 1939–1947 (2000).
  • Nosarti C , Al-AsadyMHS, FrangouS, StewartAL, RifkinL, MurrayRM. Adolescents who were born very preterm have decreased brain volumes. Brain125(7) , 1616–1623 (2002).
  • Nosarti C , GiouroukouE, HealyE et al. Grey and white matter distribution in very preterm adolescents mediates neurodevelopmental outcome. Brain 131(1) , 205–217 (2008).
  • Doesburg SM , RibaryU, HerdmanAT et al. Altered long-range phase synchronization and cortical activation in children born very preterm. IFMBE Proc. 29(9) , 250–253 (2010).
  • Doesburg SM , RibaryU, HerdmanAT et al. Magnetoencephalography reveals slowing of resting peak oscillatory frequency in children born very preterm. Pediatr. Res. 70(2) , 171–175 (2011).
  • Lax I , DuerdenE, LinS et al. Neuroanatomical consequences of very preterm birth in middle childhood. Brain Struct. Funct. 218(2) , 575–585 (2013).
  • Nosarti C , MechelliA, HerreraA et al. Structural covariance in the cortex of very preterm adolescents: a voxel-based morphometry study. Hum. Brain Mapp. 32(10) , 1615–1625 (2011).
  • Martinussen M , FischlB, LarssonHB et al. Cerebral cortex thickness in 15-year-old adolescents with low birth weight measured by an automated MRI-based method. Brain 128(Pt 11) , 2588–2596 (2005).
  • Zubiaurre-Elorza L , Soria-PastorS, JunqueC et al. Cortical thickness and behavior abnormalities in children born preterm. PLoS ONE 7(7) , e42148 (2012).
  • Nagy Z , LagercrantzH, HuttonC. Effects of preterm birth on cortical thickness measured in adolescence. Cereb. Cortex21(2) , 300–306 (2011).
  • Phillips JP , MontagueEQ, AragonM et al. Prematurity affects cortical maturation in early childhood. Pediatr. Neurol. 45(4) , 213–219 (2011).
  • Hatfield T , WingDA, BussC, HeadK, MuftulerLT, DavisEP. Magnetic resonance imaging demonstrates long-term changes in brain structure in children born preterm and exposed to chorioamnionitis. Am. J. Obstet. Gynecol.205(4) , 384–384 (2011).
  • Skranes J , L⊘haugenGC, EvensenKA et al. Entorhinal cortical thinning affects perceptual and cognitive functions in adolescents born preterm with very low birth weight (VLBW). Early Hum. Devel. 88(2) , 103–109 (2012).
  • Kesler SR , MentLR, VohrB et al. Volumetric analysis of regional cerebral development in preterm children. Pediatr. Neurol. 31(5) , 318–325 (2004).
  • Kesler SR , ReissAL, VohrB et al. Brain volume reductions within multiple cognitive systems in male preterm children at age twelve. J. Pediatri. 152(4) , 513–520 (2008).
  • Pryce CR , BettschenD, BahrNI, FeldonJ. Comparison of the effects of infant handling, isolation, and nonhandling on acoustic startle, prepulse inhibition, locomotion, and HPA activity in the adult rat. Behav. Neurosci.115(1) , 71–83 (2001).
  • Maccari S , Morley-FletcherS. Effects of prenatal restraint stress on the hypothalamus-pituitary-adrenal axis and related behavioural and neurobiological alterations. Psychoneuroendocrinology32(Suppl. 1) , S10–S15 (2007).
  • Brummelte S , PawluskiJL, GaleaLA. High post-partum levels of corticosterone given to dams influence postnatal hippocampal cell proliferation and behavior of offspring: a model of post-partum stress and possible depression. Horm. Behav.50(3) , 370–382 (2006).
  • Champagne FA , MeaneyMJ. Stress during gestation alters postpartum maternal care and the development of the offspring in a rodent model. Biol. Psychiatry59(12) , 1227–1235 (2006).
  • Bhutta AT , RovnaghiC, SimpsonPM, GossettJM, ScalzoFM, AnandKJS. Interactions of inflammatory pain and morphine in infant rats: long-term behavioral effects. Physiol. Behav.73 , 51–58 (2001).
  • Walker CD , XuZ, RochfordJ, JohnstonCC. Naturally occurring variations in maternal care modulate the effects of repeated neonatal pain on behavioral sensitivity to thermal pain in the adult offspring. Pain140(1) , 167–176 (2008).
  • de Medeiros CB , FlemingAS, JohnstonCC, WalkerCD. Artificial rearing of rat pups reveals the beneficial effects of mother care on neonatal inflammation and adult sensitivity to pain. Pediatr. Res.66(3) , 272–277 (2009).
  • Low LA , FitzgeraldM. Acute pain and a motivational pathway in adult rats: influence of early life pain experience. PLoS ONE7(3) , e34316 (2012).
  • Lupien SJ , McEwenBS, GunnarMR, HeimC. Effects of stress throughout the lifespan on the brain, behaviour and cognition. Nat. Rev. Neurosci.10(6) , 434–445 (2009).
  • McEwen BS . Physiology and neurobiology of stress and adaptation: central role of the brain. Physiol. Rev.87(3) , 873–904 (2007).
  • Anand KJ , GargS, RovnaghiCR, NarsinghaniU, BhuttaAT, HallRW. Ketamine reduces the cell death following inflammatory pain in newborn rat brain. Pediatr. Res.62(3) , 283–290 (2007).
  • Juul SE , BeyerRP, BammlerTK, FarinFM, GleasonCA. Effects of neonatal stress and morphine on murine hippocampal gene expression. Pediatr. Res.69(4) , 285–292 (2011).
  • Duhrsen L , SimonsSHP, DzietkoM et al. Effects of repetitive exposure to pain and morphine treatment on the neonatal rat brain. Neonatology 103(1) , 35–43 (2013).
  • Fabrizi L , SlaterR, WorleyA et al. A shift in sensory processing that enables the developing human brain to discriminate touch from pain. Curr. Biol. 21 , 1–7 (2011).
  • Slater R , FabriziL, WorleyA, MeekJ, BoydS, FitzgeraldM. Premature infants display increased noxious-evoked neuronal activity in the brain compared with healthy age-matched term-born infants. NeuroImage52(2) , 583–589 (2010).
  • Anand KJ . Effects of perinatal pain and stress. Prog. Brain Res.122 , 117–129 (2000).
  • Zwicker JG , GrunauRE, AdamsE et al. Score for neonatal acute physiology ii and neonatal pain predict corticospinal tract development in premature newborns. Pediatr. Neurol. 48(2) , 123–129 (2013).
  • Doesburg SM , ChauCM, CheungTPL et al. Neonatal pain-related stress, functional cortical activity and visual-perceptual abilities in school-age children born at extremely low gestational age. Pain 154(10) , 1946–1952 (2013).
  • Vinall J , MillerSP, ChauV, BrummelteS, SynnesAR, GrunauRE. Neonatal pain in relation to postnatal growth in infants born very preterm. Pain153(7) , 1374–1381 (2012).
  • Spittle AJ , TreyvaudK, DoyleLW et al. Early emergence of behavior and social-emotional problems in very preterm infants. J. Am. Acad. Child Adolesc. Psychiatry 48(9) , 909–918 (2009).
  • Vinall J , MillerSP, SynnesAR, GrunauRE. Parent behaviors moderate the relationship between neonatal pain and internalizing behaviors at 18 months corrected age in children born very prematurely. Pain154 , 1831–1839 (2013).
  • Anderson P , DoyleLW. Neurobehavioral outcomes of school-age children born extremely low birth weight or very preterm in the 1990s. JAMA289(24) , 3264–3272 (2003).
  • Loe IM , LeeES, LunaB, FeldmanHM. Behavior problems of 9–16 year old preterm children: biological, sociodemographic, and intellectual contributions. Early Hum. Dev.87(4) , 247–252 (2011).
  • Grunau RE , WhitfieldMF, FayTB. Psychosocial and academic characteristics of extremely low birth weight (< or =800 g) adolescents who are free of major impairment compared with term-born control subjects. Pediatrics114(6) , e725–e732 (2004).
  • Schmidt LA , MiskovicV, BoyleM, SaigalS. Frontal electroencephalogram asymmetry, salivary cortisol, and internalizing behavior problems in young adults who were born at extremely low birth weight. Child Dev.81(1) , 183–199 (2010).
  • van Baar AL , VermaasJ, KnotsE, de Kleine MJ, Soons P. Functioning at school age of moderately preterm children born at 32 to 36 weeks‘ gestational age. Pediatrics124(1) , 251–257 (2009).
  • Ranger M , SynnesAR, VinallJ, GrunauRE. Internalizing behaviors in school-age children born very preterm are predicted by neonatal pain and morphine exposure. Eur. J. Pain (2013) (In press).
  • Grunau RE , HaleyDW, WhitfieldMF, WeinbergJ, YuW, ThiessenP. Altered basal cortisol levels at 3, 6, 8 and 18 months in infants born at extremely low gestational age. J. Pediatr.150(2) , 151–156 (2007).
  • Grunau RE , WeinbergJ, WhitfieldMF. Neonatal procedural pain and preterm infant cortisol response to novelty at 8 months. Pediatrics114(1) , e77–e84 (2004).
  • Laprairie JL , JohnsME, MurphyAZ. Preemptive morphine analgesia attenuates the long-term consequences of neonatal inflammation in male and female rats. Pediatr. Res.64(6) , 625–630 (2008).
  • Simons SH , vanDM, van Lingen RA et al. Routine morphine infusion in preterm newborns who received ventilatory support: a randomized controlled trial. JAMA290(18) , 2419–2427 (2003).
  • Anand KJS , HallRW, DesaiN et al. Effects of morphine analgesia in ventilated preterm neonates: primary outcomes from the NEOPAIN randomised trial. Lancet 363(9422) , 1673–1682 (2004).
  • Larroque B , BreartG, KaminskiM et al. Survival of very preterm infants: epipage, a population based cohort study. Arch. Dis. Child Fetal Neonatal. Ed. 89(2) , F139–F144 (2004).
  • de Graaf J , van Lingen RA, Valkenburg AJ et al. Does neonatal morphine use affect neuropsychological outcomes at 8 to 9 years of age? Pain154(3) , 449–458 (2013).
  • de Graaf J , van Lingen RA, Simons SH et al. Long-term effects of routine morphine infusion in mechanically ventilated neonates on childrens functioning: five-year follow-up of a randomized controlled trial. Pain152(6) , 1391–1397 (2011).
  • Rozé J , DenizotS, CarbajalR et al. Prolonged sedation and/or analgesia and 5-year neurodevelopment outcome in very preterm infants: Results from the epipage cohort. Arch. Pediatr. Adolesc. Med. 162(8) , 728–733 (2008).
  • Ferguson SA , WardWL, PauleMG, HallRW, AnandKJS. A pilot study of preemptive morphine analgesia in preterm neonates: Effects on head circumference, social behavior, and response latencies in early childhood. Neurotoxicol. Teratol.34(1) , 47–55 (2012).
  • Bellu R , de Waal KA, Zanini R. Opioids for neonates receiving mechanical ventilation. Cochrane Database Syst. Rev.1 , CD004212 (2008).
  • Johnston CC , CollingeJM, HendersonSJ, AnandKJ. A cross-sectional survey of pain and pharmacological analgesia in Canadian neonatal intensive care units. Clin. J. Pain.13(4) , 308–312 (1997).
  • MacGregor R , EvansD, SugdenD, GaussenT, LeveneM. Outcome at 5–6 years of prematurely born children who received morphine as neonates. Arch. Dis. Child Fetal Neonatal Ed.79(1) , F40–F43 (1998).
  • Zwicker JG , MillerSP, GrunauRE et al. Morphine exposure is associated with altered cerebellar growth in premature newborns. Presented at: Pediatric Academic Socities Annual Meeting. Boston, MA, USA, 28 April–1 May 2012.
  • Durrmeyer X , VutskitsL, AnandKJS, RimensbergerPC. Use of analgesic and sedative drugs in the nicu: integrating clinical trials and laboratory data. Pediatr. Res.67(2) , 117–127 (2010).
  • Mason KP , LermanJ. Dexmedetomidine in children: current knowledge and future applications. Anesth. Anal.113(5) , 1129–1142 (2011).
  • Lundeberg S , StephnasonN, StillerC, EksborgS. Pharmacokinetics after a single intravenous dose of the opioid ketobemidone in neonates. Acta Anaesthesiol. Scand.56(8) , 1026–1031 (2012).
  • Cignacco E , HamersJP, StoffelL et al. The efficacy of non-pharmacological interventions in the management of procedural pain in preterm and term neonates. A systematic literature review. Eur. J. Pain. 11(2) , 139–152 (2007).
  • Campbell-Yeo M , FernandesA, JohnstonC. Procedural pain management for neonates using nonpharmacological strategies: part 2: mother-driven interventions. Adv. Neonatal. Care11(5) , 312–318 (2011).
  • Fernandes A , Campbell-YeoM, JohnstonCC. Procedural pain management for neonates using nonpharmacological strategies: part 1: Sensorial interventions. Adv. Neonatal Care11(4) , 235–241 (2011).
  • Stevens B , YamadaJ, OhlssonA. Sucrose for analgesia in newborn infants undergoing painful procedures. Cochrane Database Syst. Rev.1 , CD001069 (2010).
  • Harrison D , BeggsS, StevensB. Sucrose for procedural pain management in infants. Pediatrics130(5) , 918–925 (2012).
  • Holsti L , GrunauRE. Considerations for using sucrose to reduce procedural pain in preterm infants. Pediatrics125(5) , 1042–1047 (2010).
  • Slater R , CornelissenL, FabriziL et al. Oral sucrose as an analgesic drug for procedural pain in newborn infants: a randomised controlled trial. Lancet 376(9748) , 1225–1232 (2010).
  • Johnston CC , FilionF, SniderL et al. Routine sucrose analgesia during the first week of life in neonates younger than 31 weeks‘ postconceptional age. Pediatrics 110(3) , 523–528 (2002).
  • Walker CD . Nutritional aspects modulating brain development and the responses to stress in early neonatal life. Prog. Neuropsychopharmacol. Biol. Psychiatry29(8) , 1249–1263 (2005).
  • Spangler R , WittkowskiKM, GoddardNL, AvenaNM, HoebelBG, LeibowitzSF. Opiate-like effects of sugar on gene expression in reward areas of the rat brain. Mol. Brain Res.124(2) , 134–142 (2004).
  • Spangler R , GoddardNL, AvenaNM, HoebelBG, LeibowitzSF. Elevated D3 dopamine receptor mRNA in dopaminergic and dopaminoceptive regions of the rat brain in response to morphine. Mol. Brain Res.111 , 74–83 (2003).
  • Georges F , StinusL, BlochB, MoineCL. Chronic morphine exposure and spontaneous withdrawal are associated with modifications of dopamine receptor and neuropeptide gene expression in the rat striatum. Eur. J. Neurosci.11(2) , 481–490 (1999).
  • Colantuoni C , RadaP, McCarthyJ et al. Evidence that intermittent, excessive sugar intake causes endogenous opioid dependence. Obesity Res. 10(6) , 478–488 (2002).
  • Bello NT , LucasLR, HajnalA. Repeated sucrose access influences dopamine D2 receptor density in the striatum. Neuroreport13(12) , 1575–1578 (2002).
  • Taddio A , YiuA, SmithRW, KatzJ, McNairC, ShahV. Variability in clinical practice guidelines for sweetening agents in newborn infants undergoing painful procedures. Clin. J. Pain25(2) , 153–155 (2009).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.