1
|
Wu Y, Merhar SL, Bann CM, Newman JE, Kapse K, De Asis-Cruz J, Mack N, De Mauro SB, Ambalavanan N, Davis JM, Lorch SA, Wilson-Costello D, Poindexter BB, Peralta-Carcelen M, Limperopoulos C. Antenatal Opioid Exposure and Global and Regional Brain Volumes in Newborns. JAMA Pediatr 2025:2832261. [PMID: 40193106 PMCID: PMC11976647 DOI: 10.1001/jamapediatrics.2025.0277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/02/2025] [Indexed: 04/10/2025]
Abstract
Importance Although antenatal opioid exposure is associated with impaired brain growth, previous studies are limited by small sample sizes and lack of controls. As a result, the impacts of opioid exposure on the developing brain remain poorly understood. Objective To compare global, regional, and tissue-specific brain volumes in opioid-exposed newborns vs unexposed controls. Design, Setting, and Participants In the OBOE (Outcomes of Babies with Opioid Exposure) study, term newborns with antenatal opioid exposure and unexposed controls were recruited at 4 sites in the US from August 2020 to December 2023. Data analysis was performed from August 2020 to December 2024. Main Outcomes and Measures The primary outcome was brain volumes in both groups, assessed via unsedated 3-dimensional (3-D) volumetric magnetic resonance imaging (MRI) in opioid-exposed and unexposed newborns prior to 8 weeks of age. T2-weighted MRI data were acquired on Siemens and Philips 3T scanners and harmonized across sites. Brains were segmented using DrawEM- and 3D U-Net-based pipelines and manual corrections. Brain volumes were compared between groups using analysis of covariance, adjusting for postmenstrual age at MRI, sex, birth weight, maternal smoking, and maternal education. Results A total of 173 newborns with antenatal opioid exposure and 96 unexposed controls were studied. MRIs were performed at a mean (SD) age of 42.84 (2.11) postmenstrual weeks, and 117 newborns (43.5%) were female. The opioid-exposed group had significantly smaller total brain volume (387.51 vs 407.06 cm3; difference, 19.55; 95% CI, 8.75-30.35) and cortical (167.07 vs 176.35 cm3; difference, 9.28; 95% CI, 3.86-14.70), deep gray matter (27.22 vs 28.76 cm3; difference, 1.54; 95% CI, 0.66-2.43), white matter (159.90 vs 166.65 cm3; difference, 6.76; 95% CI, 1.71-11.81), cerebellar (23.47 vs 24.99 cm3; difference, 1.52; 95% CI, 0.67-2.36), brainstem (6.80 vs 7.18 cm3; difference, 0.38; 95% CI, 0.19-0.57), and amygdala volumes (left: 0.48 vs 0.51 cm3; difference, 0.03; 95% CI, 0.004-0.05; right: 0.51 vs 0.55 cm3; difference, 0.04; 95% CI, 0.08-0.07) compared to controls. Methadone-exposed newborns showed significantly smaller white matter volume compared to controls, while buprenorphine-exposed newborns showed significantly smaller right amygdala volume than controls. Compared to controls, newborns exposed to opioids only and those exposed to opioids plus other substances both showed significant reductions in volumes of cortical and deep gray matter, cerebellum, brainstem, right amygdala, and total brain. Polysubstance-exposed newborns additionally showed smaller volumes in white matter and the left amygdala compared to controls. Conclusions and Relevance In a large cohort of antenatally opioid-exposed newborns, there were significant reductions in global and regional brain volumes compared to unexposed controls. These data suggest vulnerability of the developing brain to antenatal opioid exposure, with varying effects depending on the type and number of substances. Trial Registration ClinicalTrials.gov Identifier: NCT04149509.
Collapse
Affiliation(s)
- Yao Wu
- Developing Brain Institute, Children’s National Hospital, Washington, DC
| | - Stephanie L. Merhar
- Perinatal Institute, Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Carla M. Bann
- Analytics Division, RTI International, Research Triangle Park, North Carolina
| | - Jamie E. Newman
- Analytics Division, RTI International, Research Triangle Park, North Carolina
| | - Kushal Kapse
- Developing Brain Institute, Children’s National Hospital, Washington, DC
| | | | - Nicole Mack
- Analytics Division, RTI International, Research Triangle Park, North Carolina
| | - Sara B. De Mauro
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- University of Pennsylvania Perelman School of Medicine, Philadelphia
| | | | | | - Scott A. Lorch
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- University of Pennsylvania Perelman School of Medicine, Philadelphia
| | | | | | | | | |
Collapse
|
2
|
Sullivan R, Ahrens Q, Mills-Huffnagle SL, Elcheva IA, Hicks SD. A human iPSC-derived midbrain neural stem cell model of prenatal opioid exposure and withdrawal: A proof of concept study. PLoS One 2025; 20:e0319418. [PMID: 40168407 PMCID: PMC11960892 DOI: 10.1371/journal.pone.0319418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/30/2025] [Indexed: 04/03/2025] Open
Abstract
A growing body of clinical literature has described neurodevelopmental delays in infants with chronic prenatal opioid exposure and withdrawal. Despite this, the mechanism of how opioids impact the developing brain remains unknown. Here, we developed an in vitro model of prenatal morphine exposure and withdrawal using healthy human induced pluripotent stem cell (iPSC)-derived midbrain neural progenitors in monolayer. To optimize our model, we identified that a longer neural induction and regional patterning period increases expression of canonical opioid receptors mu and kappa in midbrain neural progenitors compared to a shorter protocol (OPRM1, two-tailed t-test, p = 0.004; OPRK1, p = 0.0003). Next, we showed that the midbrain neural progenitors derived from a longer iPSC neural induction also have scant toll-like receptor 4 (TLR4) expression, a key player in neonatal opioid withdrawal syndrome pathophysiology. During morphine withdrawal, differentiating neural progenitors experience cyclic adenosine monophosphate overshoot compared to cell exposed to vehicle (p = 0.0496) and morphine exposure conditions (p, = 0.0136, 1-way ANOVA). Finally, we showed that morphine exposure and withdrawal alters proportions of differentiated progenitor cell fates (2-way ANOVA, F = 16.05, p < 0.0001). Chronic morphine exposure increased proportions of nestin positive progenitors (p = 0.0094), and decreased proportions of neuronal nuclear antigen positive neurons (NEUN) (p = 0.0047) compared to those exposed to vehicle. Morphine withdrawal decreased proportions of glial fibrillary acidic protein positive cells of astrocytic lineage (p = 0.044), and increased proportions of NEUN-positive neurons (p < 0.0001) compared to those exposed to morphine only. Applications of this paradigm include mechanistic studies underscoring neural progenitor cell fate commitments in early neurodevelopment during morphine exposure and withdrawal.
Collapse
MESH Headings
- Humans
- Neural Stem Cells/metabolism
- Neural Stem Cells/drug effects
- Neural Stem Cells/cytology
- Mesencephalon/cytology
- Mesencephalon/drug effects
- Mesencephalon/metabolism
- Mesencephalon/pathology
- Induced Pluripotent Stem Cells/cytology
- Induced Pluripotent Stem Cells/metabolism
- Induced Pluripotent Stem Cells/drug effects
- Morphine/adverse effects
- Morphine/pharmacology
- Female
- Analgesics, Opioid/adverse effects
- Analgesics, Opioid/pharmacology
- Cell Differentiation/drug effects
- Pregnancy
- Substance Withdrawal Syndrome/pathology
- Substance Withdrawal Syndrome/metabolism
- Prenatal Exposure Delayed Effects
- Toll-Like Receptor 4/metabolism
- Proof of Concept Study
- Receptors, Opioid, mu/metabolism
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, kappa/genetics
Collapse
Affiliation(s)
- Rhea Sullivan
- Department of Pediatrics, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Quinn Ahrens
- Department of Pediatrics, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Sara L. Mills-Huffnagle
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Irina A. Elcheva
- Department of Pediatrics, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Steven D. Hicks
- Department of Pediatrics, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| |
Collapse
|
3
|
Sahid AS, Bebbington MJ, Marcus A, Baracz SJ, Zimmermann KS, Oei J, Ward MC, Clemens KJ. Perinatal exposure to methadone or buprenorphine impairs hippocampal-dependent cognition and brain development in juvenile rats. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111255. [PMID: 39832750 DOI: 10.1016/j.pnpbp.2025.111255] [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: 07/19/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
The opioid crisis continues to escalate, disproportionately affecting women of reproductive age. Traditionally the first line of treatment for pregnant women with opioid use disorder is the mu-opioid receptor agonist methadone. However, in recent years, the use of buprenorphine as a replacement therapy has increased as it has fewer side-effects and longer duration of action. Either drug significantly improves outcomes for the mother, but their impact on the developing infant is less certain. To this end, we directly compared the effects of perinatal methadone (MET; 9 mg/kg/day starting dose) versus buprenorphine (BUP; 1 mg/kg/day starting dose) delivered via mini osmotic pump on the long-term behavior of offspring and associated molecular changes in the brain. Opioid exposure across pregnancy resulted in reduced weight gain and smaller litters compared to sham controls, and female pups in particular gained weight at a slower rate across development. Opioid treatment delayed neuromuscular reflex development, with subtle differences observed between MET and BUP. As juveniles, pups with prenatal MET exposure showed poor object recognition, although both MET and BUP have led to deficits in place recognition task. Immunofluorescence studies found corresponding decreases in astrocytes and myelin-positive cells in the hippocampus in both MET and BUP pups. Overall, both MET and BUP were associated with significant developmental and cognitive delays and changes in markers of neuronal development and inflammation, particularly in the hippocampus. The majority of changes were similar between MET and BUP-treated pups, suggesting that gestational exposure to either drug has a similar long-term negative impact on offspring.
Collapse
Affiliation(s)
- Arshman S Sahid
- School of Psychology, University of New South Wales, Sydney, Australia
| | | | - Abigail Marcus
- School of Psychology, University of New South Wales, Sydney, Australia
| | - Sarah J Baracz
- School of Psychology, University of New South Wales, Sydney, Australia
| | | | - JuLee Oei
- School of Women's and Children's Health, University of New South Wales, Kensington, NSW, Australia
| | - Meredith C Ward
- School of Women's and Children's Health, University of New South Wales, Kensington, NSW, Australia
| | - Kelly J Clemens
- School of Psychology, University of New South Wales, Sydney, Australia.
| |
Collapse
|
4
|
De Gregorio C, Gallardo J, Berríos-Cárcamo P, Handy Á, Santapau D, González-Madrid A, Ezquer M, Morales P, Luarte A, Corvalán D, Wyneken Ú, Ezquer F. Methadone directly impairs central nervous system cells in vitro. Sci Rep 2024; 14:16978. [PMID: 39043899 PMCID: PMC11266518 DOI: 10.1038/s41598-024-67860-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024] Open
Abstract
Methadone is a synthetic long-acting opioid that is increasingly used in the replacement therapy of opioid-addicted patients, including pregnant women. However, methadone therapy in this population poses challenges, as it induces cognitive and behavioral impairments in infants exposed to this opioid during prenatal development. In animal models, prenatal methadone exposure results in detrimental consequences to the central nervous system, such as: (i) increased neuronal apoptosis; (ii) disruption of oligodendrocyte maturation and increased apoptosis and (iii) increased microglia and astrocyte activation. However, it remains unclear whether these deleterious effects result from a direct effect of methadone on brain cells. Therefore, our goal was to uncover the impact of methadone on single brain cell types in vitro. Primary cultures of rat neurons, oligodendrocytes, microglia, and astrocytes were treated for three days with 10 µM methadone to emulate a chronic administration. Apoptotic neurons were identified by cleaved caspase-3 detection, and synaptic density was assessed by the juxtaposition of presynaptic and postsynaptic markers. Apoptosis of oligodendrocyte precursors was determined by cleaved caspase-3 detection. Oligodendrocyte myelination was assessed by immunofluorescence, while microglia and astrocyte proinflammatory activation were assessed by both immunofluorescence and RT-qPCR. Methadone treatment increased neuronal apoptosis and reduced synaptic density. Furthermore, it led to increased oligodendrocyte apoptosis and a reduction in the myelinating capacity of these cells, and promoted the proinflammatory activation of microglia and astrocytes. We showed that methadone, the most widely used drug in opioid replacement therapy for pregnant women with opioid addiction, directly impairs brain cells in vitro, highlighting the need for developing alternative therapies to address opioid addiction in this population.
Collapse
Affiliation(s)
| | - Javiera Gallardo
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Avenida Plaza 680, Santiago, Chile
| | - Pablo Berríos-Cárcamo
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Avenida Plaza 680, Santiago, Chile
| | - Álex Handy
- Faculty of Natural Sciences, Mathematics, and Environment, Universidad Tecnológica Metropolitana, Santiago, Chile
| | - Daniela Santapau
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Avenida Plaza 680, Santiago, Chile
| | - Antonia González-Madrid
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Avenida Plaza 680, Santiago, Chile
| | - Marcelo Ezquer
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Avenida Plaza 680, Santiago, Chile
| | - Paola Morales
- Program of Molecular and Clinical Pharmacology, ICBM, Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Alejandro Luarte
- Neuroscience Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Daniela Corvalán
- Neuroscience Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Úrsula Wyneken
- Neuroscience Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Fernando Ezquer
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Avenida Plaza 680, Santiago, Chile.
- Research Center for the Development of Novel Therapeutics Alternatives for Alcohol Use Disorders, Santiago, Chile.
| |
Collapse
|
5
|
Sorkhou M, Singla DR, Castle DJ, George TP. Birth, cognitive and behavioral effects of intrauterine cannabis exposure in infants and children: A systematic review and meta-analysis. Addiction 2024; 119:411-437. [PMID: 37968824 PMCID: PMC10872597 DOI: 10.1111/add.16370] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 09/18/2023] [Indexed: 11/17/2023]
Abstract
BACKGROUND AND AIMS Δ9-tetrahydrocannabinol (THC), the principal psychoactive component of cannabis, has been implicated in affecting fetal neurodevelopment by readily crossing the placenta. However, little is known regarding the long-term effects of intrauterine cannabis exposure. This systematic review and meta-analysis synthesized prospective and cross-sectional human studies to measure the effects of intrauterine cannabis exposure on birth, behavioral, psychological and cognitive outcomes in infancy until early childhood. METHODS Reporting according to the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) statement, cross-sectional and prospective studies published from database inception until June 2023, investigating developmental outcomes of infants, toddlers and young children with intrauterine cannabis exposure were considered. All articles were obtained from PubMed or PsycINFO databases. RESULTS The literature search resulted in 932 studies, in which 57 articles met eligibility criteria. The meta-analysis revealed that intrauterine cannabis exposure increases the risk of preterm delivery [odds ratio (OR) = 1.68, 95% confidence interval (CI) = 1.05-2.71, P = 0.03], low birth weight (OR = 2.60, CI = 1.71-3.94, P < 0.001) and requirement for neonatal intensive care unit (NICU) admission (OR = 2.51, CI = 1.46-4.31; P < 0.001). Our qualitative synthesis suggests that intrauterine cannabis exposure may be associated with poorer attention and externalizing problems in infancy and early childhood. We found no evidence for impairments in other cognitive domains or internalizing behaviors. CONCLUSIONS Prenatal cannabis use appears to be associated with lower birth weight, preterm birth and neonatal intensive care unit admission in newborns, but there is little evidence that prenatal cannabis exposure adversely impacts behavioral or cognitive outcomes in early childhood, with the exception of attention and externalizing problems.
Collapse
Affiliation(s)
- Maryam Sorkhou
- Centre for Complex Interventions, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Sciences, Department of Psychiatry, University of Toronto, ON, Canada
| | - Daisy R Singla
- Centre for Complex Interventions, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Sciences, Department of Psychiatry, University of Toronto, ON, Canada
| | - David J Castle
- Tasmania Centre for Mental Health Service Innovation, University of Tasmania, Hobart, Australia
| | - Tony P George
- Centre for Complex Interventions, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Sciences, Department of Psychiatry, University of Toronto, ON, Canada
| |
Collapse
|
6
|
Yun HJ, Nagaraj UD, Grant PE, Merhar SL, Ou X, Lin W, Acheson A, Grewen K, Kline-Fath BM, Im K. A Prospective Multi-Institutional Study Comparing the Brain Development in the Third Trimester between Opioid-Exposed and Nonexposed Fetuses Using Advanced Fetal MR Imaging Techniques. AJNR Am J Neuroradiol 2024; 45:218-223. [PMID: 38216298 PMCID: PMC11285994 DOI: 10.3174/ajnr.a8101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/07/2023] [Indexed: 01/14/2024]
Abstract
BACKGROUND AND PURPOSE While the adverse neurodevelopmental effects of prenatal opioid exposure on infants and children in the United States are well described, the underlying causative mechanisms have yet to be fully understood. This study aims to compare quantitative volumetric and surface-based features of the fetal brain between opioid-exposed fetuses and unexposed controls by using advanced MR imaging processing techniques. MATERIALS AND METHODS This is a multi-institutional IRB-approved study in which pregnant women with and without opioid use during the current pregnancy were prospectively recruited to undergo fetal MR imaging. A total of 14 opioid-exposed (31.4 ± 2.3 weeks of gestation) and 15 unexposed (31.4 ± 2.4 weeks) fetuses were included. Whole brain volume, cortical plate volume, surface area, sulcal depth, mean curvature, and gyrification index were computed as quantitative features by using our fetal brain MR imaging processing pipeline. RESULTS After correcting for gestational age, fetal sex, maternal education, polysubstance use, high blood pressure, and MR imaging acquisition site, all of the global morphologic features were significantly lower in the opioid-exposed fetuses compared with the unexposed fetuses, including brain volume, cortical volume, cortical surface area, sulcal depth, cortical mean curvature, and gyrification index. In regional analysis, the opioid-exposed fetuses showed significantly decreased surface area and sulcal depth in the bilateral Sylvian fissures, central sulci, parieto-occipital fissures, temporal cortices, and frontal cortices. CONCLUSIONS In this small cohort, prenatal opioid exposure was associated with altered fetal brain development in the third trimester. This adds to the growing body of literature demonstrating that prenatal opioid exposure affects the developing brain.
Collapse
Affiliation(s)
- Hyuk Jin Yun
- From the Division of Newborn Medicine (H.J.Y, P.E.G., K.I.), Boston Children's Hospital, Boston, MA
- Harvard Medical School (H.J.Y, P.E.G., K.I.), Boston, MA
| | - Usha D Nagaraj
- Department of Radiology and Medical Imaging (U.D.N., B.M.K.-F.), Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine (U.D.N., S.L.M., B.M.K.-F.), Cincinnati, OH
| | - P Ellen Grant
- From the Division of Newborn Medicine (H.J.Y, P.E.G., K.I.), Boston Children's Hospital, Boston, MA
- Harvard Medical School (H.J.Y, P.E.G., K.I.), Boston, MA
- Department of Radiology (P.E.G.), Boston Children's Hospital, Boston, MA
| | - Stephanie L Merhar
- University of Cincinnati College of Medicine (U.D.N., S.L.M., B.M.K.-F.), Cincinnati, OH
- Division of Neonatology, Perinatal Institute (S.L.M.), Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Xiawei Ou
- Departments of Radiology and Pediatrics (X.O.), University of Arkansas for Medical Sciences, Little Rock, AR
| | - Weili Lin
- Department of Radiology (W.L.), University of North Carolina, Chappel Hill, NC
| | - Ashley Acheson
- Department of Psychiatry and Behavioral Sciences (A.A.), University of Arkansas for Medical Sciences, Little Rock, AR
| | - Karen Grewen
- Department of Psychiatry (K.G.), University of North Carolina, Chappel Hill, NC
| | - Beth M Kline-Fath
- Department of Radiology and Medical Imaging (U.D.N., B.M.K.-F.), Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine (U.D.N., S.L.M., B.M.K.-F.), Cincinnati, OH
| | - Kiho Im
- From the Division of Newborn Medicine (H.J.Y, P.E.G., K.I.), Boston Children's Hospital, Boston, MA
- Harvard Medical School (H.J.Y, P.E.G., K.I.), Boston, MA
| |
Collapse
|
7
|
Dudley JA, Nagaraj UD, Merhar S, Mangano FT, Kline-Fath BM, Ou X, Acheson A, Yuan W. DTI of Opioid-Exposed Fetuses Using ComBat Harmonization: A Bi-Institutional Study. AJNR Am J Neuroradiol 2023; 44:1084-1089. [PMID: 37562830 PMCID: PMC10494946 DOI: 10.3174/ajnr.a7951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/25/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND AND PURPOSE The underlying mechanisms leading to altered cognitive, behavioral, and vision outcomes in children with prenatal opioid exposure are yet to be fully understood. Some studies suggest WM alterations in infants and children with prenatal opioid exposure; however, the time course of WM changes is unknown. We aimed to evaluate differences in diffusion tensor imaging MRI parameters in the brain between opioid exposed fetuses and normal controls. MATERIALS AND METHODS This is a pilot, prospective cohort study in which subjects in the third trimester of pregnancy underwent fetal DTI of the brain with 20 noncolinear diffusion directions and a b-value of 500 s/mm2 at 2.5-mm isotropic resolution. RESULTS The study included a total of 26 fetuses, 11 opioid-exposed (mean gestational age, 32.61 [SD, 2.35] weeks) and 15 unexposed controls (mean gestational age, 31.77 [SD, 1.68] weeks). After we adjusted for gestational age, fractional anisotropy values were significantly higher in opioid-exposed fetuses relative to controls in 8 WM tracts: the bilateral lemniscus (left: P = .017; right: P = .020), middle cerebellar peduncle (P = .027), left inferior cerebellar peduncle (P = .026), right sagittal stratum (P = .040), right fornix stria terminalis (P = .022), right inferior fronto-occipital fasciculus (P = .011), and the right uncinate fasciculus (P = .033). Significant alteration was also identified in other DTI indices involving a series of brain regions. CONCLUSIONS Our data demonstrate initial evidence of cerebral WM microstructural differences between opioid-exposed fetuses and unexposed controls. Further studies in larger patient populations will be needed to fully understand these findings.
Collapse
Affiliation(s)
- J A Dudley
- From the Department of Radiology and Medical Imaging (J.A.D., U.D.N., B.M.K.-F., W.Y.), Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- University of Cincinnati College of Medicine (J.A.D., U.D.N., S.M., F.T.M., B.M.K.-F., W.Y.), Cincinnati, Ohio
| | - U D Nagaraj
- From the Department of Radiology and Medical Imaging (J.A.D., U.D.N., B.M.K.-F., W.Y.), Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- University of Cincinnati College of Medicine (J.A.D., U.D.N., S.M., F.T.M., B.M.K.-F., W.Y.), Cincinnati, Ohio
| | - S Merhar
- University of Cincinnati College of Medicine (J.A.D., U.D.N., S.M., F.T.M., B.M.K.-F., W.Y.), Cincinnati, Ohio
- Perinatal Institute, Division of Neonatology (S.M.), Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - F T Mangano
- University of Cincinnati College of Medicine (J.A.D., U.D.N., S.M., F.T.M., B.M.K.-F., W.Y.), Cincinnati, Ohio
- Department of Neurosurgery (F.T.M.), Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - B M Kline-Fath
- From the Department of Radiology and Medical Imaging (J.A.D., U.D.N., B.M.K.-F., W.Y.), Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- University of Cincinnati College of Medicine (J.A.D., U.D.N., S.M., F.T.M., B.M.K.-F., W.Y.), Cincinnati, Ohio
| | - X Ou
- Departments of Radiology (X.O.), University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Departments of Pediatrics (X.O.), University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - A Acheson
- Department of Psychiatry (A.A.), University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - W Yuan
- From the Department of Radiology and Medical Imaging (J.A.D., U.D.N., B.M.K.-F., W.Y.), Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- University of Cincinnati College of Medicine (J.A.D., U.D.N., S.M., F.T.M., B.M.K.-F., W.Y.), Cincinnati, Ohio
| |
Collapse
|
8
|
Chin EM, Kitase Y, Madurai NK, Robinson S, Jantzie LL. In utero methadone exposure permanently alters anatomical and functional connectivity: A preclinical evaluation. Front Pediatr 2023; 11:1139378. [PMID: 36911026 PMCID: PMC9995894 DOI: 10.3389/fped.2023.1139378] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/03/2023] [Indexed: 02/25/2023] Open
Abstract
The opioid epidemic is an ongoing public health crisis, and children born following prenatal opioid exposure (POE) have increased risk of long-term cognitive and behavioral sequelae. Clinical studies have identified reduced gray matter volume and abnormal white matter microstructure in children with POE but impacts on whole-brain functional brain connectivity (FC) have not been reported. To define effects of POE on whole brain FC and white matter injury in adult animals, we performed quantitative whole-brain structural and functional MRI. We used an established rat model of POE in which we have previously reported impaired executive function in adult rats analogous to persistent neurocognitive symptoms described in humans with POE. Pregnant Sprague-Dawley rat dams received continuous methadone (12 mg/kg/day) vs. saline infusion for 28 days via osmotic mini-pumps, exposing rats to pre- and postnatal opioid until weaning. At young adult age (P60), POE and saline exposed offspring underwent in vivo MRI included diffusion tensor imaging and functional MRI (fMRI). Results indicate that fractional anisotropy (FA) was decreased in adult animals with POE [n = 11] compared to animals that received saline [n = 9] in major white matter tracts, including the corpus callosum (p < 0.001) and external capsule (p < 0.01). This change in FA was concomitant with reduced axial diffusivity in the external capsule (p < 0.01) and increased radial diffusivity in the corpus callosum (p < 0.01). fMRI analyses reveal brainwide FC was diffusely lower in POE (p < 10-6; 10% of variance explained by group). Decreased connectivity in cortical-cortical and cortico-basal ganglia circuitry was particularly prominent with large effect sizes (Glass's Δ > 1). Taken together, these data confirm POE reduces brainwide functional connectivity as well as microstructural integrity of major white matter tracts. Altered neural circuitry, dysregulated network refinement, and diffuse network dysfunction have been implicated in executive function deficits that are common in children with POE. FC may serve as a translatable biomarker in children with POE.
Collapse
Affiliation(s)
- Eric M. Chin
- Department of Neurodevelopmental Medicine, Phelps Center for Cerebral Palsy and Neurodevelopmental Medicine, Kennedy Krieger Institute, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yuma Kitase
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nethra K. Madurai
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shenandoah Robinson
- Department of Neurodevelopmental Medicine, Phelps Center for Cerebral Palsy and Neurodevelopmental Medicine, Kennedy Krieger Institute, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lauren L. Jantzie
- Department of Neurodevelopmental Medicine, Phelps Center for Cerebral Palsy and Neurodevelopmental Medicine, Kennedy Krieger Institute, Baltimore, MD, United States
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
9
|
Balalian AA, Graeve R, Richter M, Fink A, Kielstein H, Martins SS, Philbin MM, Factor-Litvak P. Prenatal exposure to opioids and neurodevelopment in infancy and childhood: A systematic review. Front Pediatr 2023; 11:1071889. [PMID: 36896405 PMCID: PMC9989202 DOI: 10.3389/fped.2023.1071889] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/23/2023] [Indexed: 02/23/2023] Open
Abstract
Aim This systematic review aims to estimate the relationship between prenatal exposure to opioids and neurodevelopmental outcomes and examines potential sources of heterogeneity between the studies. Methods We searched four databases through May 21st, 2022: PubMed, Embase, PsycInfo and the Web of Science according to a specified search strings. Study inclusion criteria include: (1) cohort and case-control peer-reviewed studies published in English; (2) studies comparing neurodevelopmental outcomes among children with prenatal opioid-exposure (prescribed or used non-medically) vs. an unexposed group. Studies investigating fetal alcohol syndrome or a different primary prenatal exposure other than opioids were excluded. Two main performed data extraction using "Covidence" systematic review platform. This systematic review was conducted in accordance with PRISMA guidelines. The Newcastle-Ottawa-Scale was used for quality assessment of the studies. Studies were synthesized based on the type of neurodevelopmental outcome and the instrument used to assess neurodevelopment. Results Data were extracted from 79 studies. We found significant heterogeneity between studies due to their use of different instruments to explore cognitive skills, motor, and behavioral outcomes among children of different ages. The other sources of heterogeneity included: procedures to assess prenatal exposure to opioids; period of pregnancy in which exposure was assessed; type of opioids assessed (non-medical, medication used for opioid use dis-order, prescribed by health professional), types of co-exposure; source of selection of prenatally exposed study participants and comparison groups; and methods to address lack of comparability between exposed and unexposed groups. Cognitive and motor skills as well as behavior were generally negatively affected by prenatal opioid exposure, but the significant heterogeneity precluded a meta-analysis. Conclusion We explored sources of heterogeneity in the studies assessing the association between prenatal exposure to opioids and neurodevelopmental outcomes. Sources of heterogeneity included different approaches to participant recruitment as well as exposure and outcome ascertainment methods. Nonetheless, overall negative trends were observed between prenatal opioid exposure and neuro-developmental outcomes.
Collapse
Affiliation(s)
- Arin A. Balalian
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Richard Graeve
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Matthias Richter
- Social Determinants of Health Group, Department of Sport and Health Sciences, Technical University of Munich (TUM), Germany
| | - Astrid Fink
- Department of Health and Consumer Protection, Kreis Groß-Gerau, Groß-Gerau, Germany
| | - Heike Kielstein
- Institut für Anatomie und Zellbiologie, Martin-Luther-Universität Halle, Halle (Saale), Germany
| | - Silvia S. Martins
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Morgan M. Philbin
- Department of Sociomedical Sciences, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Pam Factor-Litvak
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, United States
| |
Collapse
|
10
|
Hornburg KJ, Slosky LM, Cofer G, Cook J, Qi Y, Porkka F, Clark NB, Pires A, Petrella JR, White LE, Wetsel WC, Barak L, Caron MG, Johnson GA. Prenatal heroin exposure alters brain morphology and connectivity in adolescent mice. NMR IN BIOMEDICINE 2023; 36:e4842. [PMID: 36259728 PMCID: PMC10483958 DOI: 10.1002/nbm.4842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/30/2022] [Accepted: 10/05/2022] [Indexed: 06/16/2023]
Abstract
The United States is experiencing a dramatic increase in maternal opioid misuse and, consequently, the number of individuals exposed to opioids in utero. Prenatal opioid exposure has both acute and long-lasting effects on health and wellbeing. Effects on the brain, often identified at school age, manifest as cognitive impairment, attention deficit, and reduced scholastic achievement. The neurobiological basis for these effects is poorly understood. Here, we examine how in utero exposure to heroin affects brain development into early adolescence in a mouse model. Pregnant C57BL/6J mice received escalating doses of heroin twice daily on gestational days 4-18. The brains of offspring were assessed on postnatal day 28 using 9.4 T diffusion MRI of postmortem specimens at 36 μm resolution. Whole-brain volumes and the volumes of 166 bilateral regions were compared between heroin-exposed and control offspring. We identified a reduction in whole-brain volume in heroin-exposed offspring and heroin-associated volume changes in 29 regions after standardizing for whole-brain volume. Regions with bilaterally reduced standardized volumes in heroin-exposed offspring relative to controls include the ectorhinal and insular cortices. Regions with bilaterally increased standardized volumes in heroin-exposed offspring relative to controls include the periaqueductal gray, septal region, striatum, and hypothalamus. Leveraging microscopic resolution diffusion tensor imaging and precise regional parcellation, we generated whole-brain structural MRI diffusion connectomes. Using a dimension reduction approach with multivariate analysis of variance to assess group differences in the connectome, we found that in utero heroin exposure altered structure-based connectivity of the left septal region and the region that acts as a hub for limbic regulatory actions. Consistent with clinical evidence, our findings suggest that prenatal opioid exposure may have effects on brain morphology, connectivity, and, consequently, function that persist into adolescence. This work expands our understanding of the risks associated with opioid misuse during pregnancy and identifies biomarkers that may facilitate diagnosis and treatment.
Collapse
Affiliation(s)
- Kathryn J. Hornburg
- Department of Radiology, School of Medicine, Duke University; 311 Research Drive; Campus Box 3302; Durham, NC 27710 United States
| | - Lauren M. Slosky
- Department of Cell Biology, School of Medicine, Duke University; Campus Box 3709; Durham, NC 27710 United States
- Department of Pharmacology, University of Minnesota; 312 Church Street SE; 3-104 Nils Hasselmo Hall; Minneapolis, MN 55455 United States
| | - Gary Cofer
- Department of Radiology, School of Medicine, Duke University; 311 Research Drive; Campus Box 3302; Durham, NC 27710 United States
| | - James Cook
- Department of Radiology, School of Medicine, Duke University; 311 Research Drive; Campus Box 3302; Durham, NC 27710 United States
| | - Yi Qi
- Department of Radiology, School of Medicine, Duke University; 311 Research Drive; Campus Box 3302; Durham, NC 27710 United States
| | - Fiona Porkka
- Department of Cell Biology, School of Medicine, Duke University; Campus Box 3709; Durham, NC 27710 United States
| | - Nicholas B. Clark
- Department of Cell Biology, School of Medicine, Duke University; Campus Box 3709; Durham, NC 27710 United States
| | - Andrea Pires
- Department of Cell Biology, School of Medicine, Duke University; Campus Box 3709; Durham, NC 27710 United States
| | - Jeffrey R Petrella
- Department of Radiology, School of Medicine, Duke University; 311 Research Drive; Campus Box 3302; Durham, NC 27710 United States
| | - Leonard E. White
- Department of Neurology, School of Medicine, Duke University; Campus Box 2900; Durham, NC 27710 United States
| | - William C. Wetsel
- Department of Cell Biology, School of Medicine, Duke University; Campus Box 3709; Durham, NC 27710 United States
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Duke University; Campus Box 102508; Durham, NC 27710 United States
- Department of Neurology, School of Medicine, Duke University; Campus Box 2900; Durham, NC 27710 United States
| | - Lawrence Barak
- Department of Cell Biology, School of Medicine, Duke University; Campus Box 3709; Durham, NC 27710 United States
| | - Marc G. Caron
- Department of Cell Biology, School of Medicine, Duke University; Campus Box 3709; Durham, NC 27710 United States
- Department of Neurology, School of Medicine, Duke University; Campus Box 2900; Durham, NC 27710 United States
| | - G. Allan Johnson
- Department of Radiology, School of Medicine, Duke University; 311 Research Drive; Campus Box 3302; Durham, NC 27710 United States
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University; Campus Box 90281; Durham, NC 27708-0281 United States
| |
Collapse
|
11
|
Madurai NK, Kitase Y, Hamimi S, Kirk SE, Sevensky R, Ramachandra S, Muthukumar S, Vasan V, Ozen M, Gerner G, Robinson S, Jantzie LL. Methadone alters the peripheral inflammatory and central immune landscape following prenatal exposure in rats. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2022; 2:10792. [PMID: 37396628 PMCID: PMC10312988 DOI: 10.3389/adar.2022.10792] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Opioid use during pregnancy continues to rise at alarming rates with a parallel trend in the number of infants and children exposed to opioid medications each year. Prenatal opioid exposure (POE) occurs at a critical timepoint in neurodevelopment disrupting intricate pathways essential for neural-immune maturation with the potential for devastating long-term consequences. Understanding the mechanisms underlying injury associated with POE is essential to address long-term outcomes and identify diagnostic and therapeutic biomarkers in this vulnerable patient population. Using an established preclinical model of POE, we investigated changes in cerebral and peripheral inflammation and peripheral blood mononuclear cell (PBMC) activity. We hypothesized that neuroinflammation, as defined by changes in specific cerebral immune cell populations, would exist in adult rats following POE concomitant with sustained peripheral immune hyperreactivity (SPIHR). Our data demonstrated alterations in cerebral immune cells at postnatal day 60 (P60) typified by increased regulatory T cells (p < 0.01) and neutrophils (p < 0.05) in rats with POE compared to controls. Evaluation of serum revealed increased levels of IL-6 (p < 0.05) and CXCL1 (p < 0.05) at P21 in rats with POE compared to controls with no significant difference in cytokine or chemokine levels between the two groups at P60. Additionally, PBMCs isolated from rats with POE at P21 demonstrated baseline hypersecretion of IL-6 (p < 0.01) and SPIHR with increased levels of TNF-α (p < 0.05) and CXCL1 (p < 0.05) following stimulation with LPS. At P60, however, there was no significant difference found in cytokine or chemokine levels secreted by PBMCs isolated from rats with POE at baseline or with LPS stimulation when compared to controls. Taken together, these data demonstrate cerebral inflammation months after prenatal opioid exposure and long after the resolution of systemic inflammation and SPIHR seen at toddler age equivalent. Chronic alterations in the cerebral immune cell populations secondary to prenatal opioid exposure may underly long-term consequences of developmental brain injury including deficits in cognition and attention. These findings may be invaluable to further investigations of precise biomarkers of injury and targeted therapeutics for this vulnerable population.
Collapse
Affiliation(s)
- Nethra K. Madurai
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Yuma Kitase
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Sarah Hamimi
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
- Division of Pediatric Neurosurgery, Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Shannon E. Kirk
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Riley Sevensky
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Sindhu Ramachandra
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Sankar Muthukumar
- Division of Pediatric Neurosurgery, Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Vikram Vasan
- Division of Pediatric Neurosurgery, Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Maide Ozen
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Gwendolyn Gerner
- Department of Neuropsychology, Kennedy Krieger Institute, Baltimore, MD, United States
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Shenandoah Robinson
- Division of Pediatric Neurosurgery, Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Lauren L. Jantzie
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
- Division of Pediatric Neurosurgery, Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
- Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, MD, United States
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
12
|
Grecco GG, Shahid SS, Atwood BK, Wu YC. Alterations of brain microstructures in a mouse model of prenatal opioid exposure detected by diffusion MRI. Sci Rep 2022; 12:17085. [PMID: 36224335 PMCID: PMC9556691 DOI: 10.1038/s41598-022-21416-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 09/27/2022] [Indexed: 01/04/2023] Open
Abstract
Growing opioid use among pregnant women is fueling a crisis of infants born with prenatal opioid exposure. A large body of research has been devoted to studying the management of opioid withdrawal during the neonatal period in these infants, but less substantive work has explored the long-term impact of prenatal opioid exposure on neurodevelopment. Using a translationally relevant mouse model of prenatal methadone exposure (PME), the aim of the study is to investigate the cerebral microstructural differences between the mice with PME and prenatal saline exposure (PSE). The brains of eight-week-old male offspring with either PME (n = 15) or PSE (n = 15) were imaged using high resolution in-vivo diffusion magnetic resonance imaging on a 9.4 Tesla small animal scanner. Brain microstructure was characterized using diffusion tensor imaging (DTI) and Bingham neurite orientation dispersion and density imaging (Bingham-NODDI). Voxel-based analysis (VBA) was performed using the calculated microstructural parametric maps. The VBA showed significant (p < 0.05) bilateral alterations in fractional anisotropy (FA), mean diffusivity (MD), axial diffusivity (AD), radial diffusivity (RD), orientation dispersion index (ODI) and dispersion anisotropy index (DAI) across several cortical and subcortical regions, compared to PSE. Particularly, in PME offspring, FA, MD and AD were significantly higher in the hippocampus, dorsal amygdala, thalamus, septal nuclei, dorsal striatum and nucleus accumbens. These DTI-based results suggest widespread bilateral microstructural alterations across cortical and subcortical regions in PME offspring. Consistent with the observations in DTI, Bingham-NODDI derived ODI exhibited significant reduction in PME offspring within the hippocampus, dorsal striatum and cortex. NODDI-based results further suggest reduction in dendritic arborization in PME offspring across multiple cortical and subcortical regions. To our best knowledge, this is the first study of prenatal opioid exposure to examine microstructural organization in vivo. Our findings demonstrate perturbed microstructural complexity in cortical and subcortical regions persisting into early adulthood which could interfere with critical neurodevelopmental processes in individuals with prenatal opioid exposure.
Collapse
Affiliation(s)
- Gregory G Grecco
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana University School of Medicine, Medical Scientist Training Program, Indianapolis, IN, 46202, USA
| | - Syed Salman Shahid
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 West 16th Street, Suite 4100, Indianapolis, IN, 46202, USA
| | - Brady K Atwood
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yu-Chien Wu
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 West 16th Street, Suite 4100, Indianapolis, IN, 46202, USA.
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
13
|
Vishnubhotla RV, Zhao Y, Wen Q, Dietrich J, Sokol GM, Sadhasivam S, Radhakrishnan R. Brain structural connectome in neonates with prenatal opioid exposure. Front Neurosci 2022; 16:952322. [PMID: 36188457 PMCID: PMC9523134 DOI: 10.3389/fnins.2022.952322] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
IntroductionInfants with prenatal opioid exposure (POE) are shown to be at risk for poor long-term neurobehavioral and cognitive outcomes. Early detection of brain developmental alterations on neuroimaging could help in understanding the effect of opioids on the developing brain. Recent studies have shown altered brain functional network connectivity through the application of graph theoretical modeling, in infants with POE. In this study, we assess global brain structural connectivity through diffusion tensor imaging (DTI) metrics and apply graph theoretical modeling to brain structural connectivity in infants with POE.MethodsIn this prospective observational study in infants with POE and control infants, brain MRI including DTI was performed before completion of 3 months corrected postmenstrual age. Tractography was performed on the whole brain using a deterministic fiber tracking algorithm. Pairwise connectivity and network measure were calculated based on fiber count and fractional anisotropy (FA) values. Graph theoretical metrics were also derived.ResultsThere were 11 POE and 18 unexposed infants included in the analysis. Pairwise connectivity based on fiber count showed alterations in 32 connections. Pairwise connectivity based on FA values showed alterations in 24 connections. Connections between the right superior frontal gyrus and right paracentral lobule and between the right superior occipital gyrus and right fusiform gyrus were significantly different after adjusting for multiple comparisons between POE infants and unexposed controls. Additionally, alterations in graph theoretical network metrics were identified with fiber count and FA value derived tracts.ConclusionComparisons show significant differences in fiber count in two structural connections. The long-term clinical outcomes related to these findings may be assessed in longitudinal follow-up studies.
Collapse
Affiliation(s)
- Ramana V. Vishnubhotla
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Yi Zhao
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Qiuting Wen
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jonathan Dietrich
- Indiana University School of Medicine, Indianapolis, IN, United States
| | - Gregory M. Sokol
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Senthilkumar Sadhasivam
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Rupa Radhakrishnan
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
- *Correspondence: Rupa Radhakrishnan,
| |
Collapse
|
14
|
Lowell AF, Morie K, Potenza MN, Crowley MJ, Mayes LC. An intergenerational lifespan perspective on the neuroscience of prenatal substance exposure. Pharmacol Biochem Behav 2022; 219:173445. [PMID: 35970340 DOI: 10.1016/j.pbb.2022.173445] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 07/26/2022] [Accepted: 08/05/2022] [Indexed: 11/16/2022]
Abstract
Prenatal substance exposure has the potential to impact a variety of domains, with neurobiological effects that last throughout the lifespan. Different substances may impact the brain in both specific and diffuse ways; however, the aberrant neural outcomes following exposure tend to coalesce in three areas: (1) sensorimotor development; (2) arousal, motivation, and reward; and (3) executive functioning, impulse control, and emotion regulation. This manuscript represents a summary and update of a previous review (Morie et al., 2019). We organize this piece by domain and summarize data from published neuroimaging studies that examine the neural correlates of prenatal exposure across developmental stages. While the published neuroimaging literature in the area of prenatal exposure has a range of sampling concerns that may limit generalizability as well as longitudinal prediction, the findings to date do point to domains of interest warranting further study. With this caveat, we synthesize the extant findings to describe ways in which prenatal substance exposure is associated with developmental psychopathology and implicated in potentially aberrant behavioral patterns beginning in infancy and persisting through childhood, adolescence, adulthood, and even parenthood. We also examine how substance abuse may impact parenting behaviors that in turn influences infant and child behavior in ways that may be additive or obscure the direct teratological effects of prenatal exposure. Given this observation, we offer an additional intergenerational lens through which prenatal substance exposure should be studied.
Collapse
Affiliation(s)
- Amanda F Lowell
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, USA.
| | - Kristen Morie
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Marc N Potenza
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA; Connecticut Mental Health Center, New Haven, CT, USA; Connecticut Council on Problem Gambling, Wethersfield, CT, USA
| | - Michael J Crowley
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Linda C Mayes
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
15
|
Gibson JM, Chu T, Zeng W, Wethall AC, Kong M, Mellen N, Devlin Phinney LA, Cai J. Perinatal methadone exposure attenuates myelination and induces oligodendrocyte apoptosis in neonatal rat brain. Exp Biol Med (Maywood) 2022; 247:1067-1079. [PMID: 35475383 PMCID: PMC9265527 DOI: 10.1177/15353702221090457] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 03/09/2022] [Indexed: 11/15/2022] Open
Abstract
Methadone (MTD) is a commonly prescribed treatment for opioid use disorder in pregnancy, despite limited information on the effects of passive exposure on fetal brain development. Animal studies suggest a link between perinatal MTD exposure and impaired white matter development. In this study, we characterized the effect of perinatal MTD exposure through the evaluation of oligodendrocyte development and glial cell activation in the neonatal rat brain. Six pregnant Sprague Dawley rat dams were randomized to MTD (0.2 mL/L) or untreated drinking water from embryonic day 7. Pups were terminated at postnatal day 7 and tissue sections were harvested from six randomly selected pups (one male and one female per litter) of each experimental group for immunohistochemistry in areas of corpus callosum (CC), lateral CC, external capsule (EC), and cerebellar white matter. In the MTD-exposed rat pups, myelination was significantly decreased in the CC, lateral CC, EC, and arbor vitae compared with the controls. The increased density and percentage of oligodendrocyte precursor cells (OPCs) were observed in the CC and cerebellar white matter. The highly active proliferation of OPCs as well as decreased density and percentage of differentiated oligodendrocytes were found in the cerebellum but no differences in the cerebrum. Apoptotic activities of both differentiated oligodendrocytes and myelinating oligodendrocytes were significantly increased in all regions of the cerebrum and cerebellum after MTD exposure. There was no quantitative difference in astrocyte, however, cell density and/or morphologic difference consistent with activation were observed in microglia throughout MTD-exposed CC and cerebellum. Taken together, perinatal MTD exposure reveals global attenuation of myelination, accelerated apoptosis of both differentiated and myelinating oligodendrocytes, and microglia activation, supporting an association between antenatal MTD exposure and impaired myelination in the developing brain.
Collapse
Affiliation(s)
- Jennifer M Gibson
- Division of Neonatology, Department of Pediatrics, School of Medicine, University of Louisville, Norton Children’s Hospital, Norton Healthcare, Louisville, KY 40202, USA
| | - Tianci Chu
- Pediatric Research Institute, Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Wenxin Zeng
- Pediatric Research Institute, Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Ashley C Wethall
- Division of Neonatology, Department of Pediatrics, School of Medicine, University of Louisville, Norton Children’s Hospital, Norton Healthcare, Louisville, KY 40202, USA
- Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Maiying Kong
- Department of Bioinformatics and Biostatistics, School of Public Health & Information Sciences, University of Louisville, Louisville, KY 40202, USA
| | - Nicholas Mellen
- Department of Neurology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Lori A Devlin Phinney
- Division of Neonatology, Department of Pediatrics, School of Medicine, University of Louisville, Norton Children’s Hospital, Norton Healthcare, Louisville, KY 40202, USA
| | - Jun Cai
- Pediatric Research Institute, Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
16
|
Sithisarn T, Legan SJ, Westgate PM, Bada HS, Wilson ME. Understanding the effects of opioids vs non-opioids in the treatment of neonatal abstinence syndrome, an in vitro model. Front Pediatr 2022; 10:1068330. [PMID: 36483468 PMCID: PMC9723166 DOI: 10.3389/fped.2022.1068330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/04/2022] [Indexed: 11/23/2022] Open
Abstract
Neonatal abstinence syndrome (NAS) refers to cadre of withdrawal manifestations in infants born to mothers who used illicit and licit substances during pregnancy. The increasing prevalence of NAS has been largely due to the maternal use of opioids during pregnancy. NAS contributes to increased morbidity and long-term disability in surviving infants. Clinically, oral opioid therapies for opioid exposure have been a standard treatment with morphine (MO) being the most commonly used medication. Recently, a non-opioid agent, clonidine (CD) has also been used with potentially favorable short- and long-term outcomes in infants. However, data regarding the cellular and molecular effects of these treatments on the developing brain is still lacking due to a lack of a reliable animal model that targets the neonatal brain. To address this gap in knowledge we determined the effects of MO or CD on the cell death of neonatal cortical explant cultures that were exposed to oxycodone (OXY) in utero. Sprague Dawley rats were randomized and implanted with programmable infusion pumps before mating to receive either the OXY (dose increasing from 1.21-1.90 mg/kg/day to a maximum dose of 2.86-3.49 mg/kg/day) or normal saline (NS) throughout pregnancy and until one week after delivery. Male and female rat pups were sacrificed on postnatal day 4, and the prefrontal cortex (PFC) and hippocampus (HC) were dissected and treated with MO (0.10-1.00 µM) or CD (1.20-120.00 µM) in culture media. After 5 days of treatment the explants were labeled with propidium iodide to detect cell death. Dead cells were analyzed and counted under fluorescence microscopy. In explants from the PFC, cell death was greater in those prenatally exposed to OXY and postnatally treated with MO (OXY/MO) (736.8 ± 76.5) compared to OXY/CD (620.9 ± 75.0; p = 0.005). In the HC explants, mean cell death counts were not significantly different between groups regardless of prenatal exposure or postnatal treatment (p = 0.19). The PFC is vital in controlling higher-order executive functions such as behavioral flexibility, learning and working memory. Therefore, our finding is consistent with executive function problems in children with prenatal opioid exposure.
Collapse
Affiliation(s)
- Thitinart Sithisarn
- Department of Pediatrics, University of Kentucky, Lexington KY, United States
| | - Sandra J Legan
- Department of Physiology, University of Kentucky, Lexington KY, United States
| | - Philip M Westgate
- Department of Biostatistics, University of Kentucky, Lexington KY, United States
| | - Henrietta S Bada
- Department of Pediatrics, University of Kentucky, Lexington KY, United States
| | - Melinda E Wilson
- Department of Physiology, University of Kentucky, Lexington KY, United States
| |
Collapse
|
17
|
Vassoler FM, Wimmer ME. Consequences of Parental Opioid Exposure on Neurophysiology, Behavior, and Health in the Next Generations. Cold Spring Harb Perspect Med 2021; 11:a040436. [PMID: 32601130 PMCID: PMC8485740 DOI: 10.1101/cshperspect.a040436] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Substance abuse and the ongoing opioid epidemic represents a large societal burden. This review will consider the long-term impact of opioid exposure on future generations. Prenatal, perinatal, and preconception exposure are reviewed with discussion of both maternal and paternal influences. Opioid exposure can have long-lasting effects on reproductive function, gametogenesis, and germline epigenetic programming, which can influence embryogenesis and alter the developmental trajectory of progeny. The potential mechanisms by which preconception maternal and paternal opioid exposure produce deleterious consequences on the health, behavior, and physiology of offspring that have been identified by clinical and animal studies will be discussed. The timing, nature, dosing, and duration of prenatal opioid exposure combined with other important environmental considerations influence the extent to which these manipulations affect parents and their progeny. Epigenetic inheritance refers to the transmission of environmental insults across generations via mechanisms independent of the DNA sequence. This topic will be further explored in the context of prenatal, perinatal, and preconception opioid exposure for both the maternal and paternal lineage.
Collapse
Affiliation(s)
- Fair M Vassoler
- Tufts University, Cummings School of Veterinary Medicine, Grafton, Massachusetts 01536, USA
| | - Mathieu E Wimmer
- Department of Psychology and Program in Neuroscience, Temple University, Philadelphia, Pennsylvania 19122, USA
| |
Collapse
|
18
|
Yazdanfar N, Farnam A, Sadigh-Eteghad S, Mahmoudi J, Sarkaki A. Enriched environment and social isolation differentially modulate addiction-related behaviors in male offspring of morphine-addicted dams: The possible role of μ-opioid receptors and ΔFosB in the brain reward pathway. Brain Res Bull 2021; 170:98-105. [PMID: 33592274 DOI: 10.1016/j.brainresbull.2021.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 01/19/2021] [Accepted: 02/02/2021] [Indexed: 12/20/2022]
Abstract
Prenatal opioids exposure negatively affects the neurobehavioral abilities of children born from dependence dams. Adolescent housing conditions can buffer the detrimental impacts of early life experiences or contradictory can worsen individual psychosocial functions. The present study investigated the effects of maternal morphine dependence and different rearing conditions on behaviors and protein expression in brain reward circuits of male pups. Female Wistar rats a week before conception, during pregnancy and lactation were injected twice daily with escalating doses of morphine or saline. On a postnatal day 21, male pups were weaned and subjected to three different environments for two months: standard (STD), isolated (ISO), or enriched environment (EE). The anxiety and drug-related reward were measured using elevated plus maze, open field test, and conditioned place preference. Western blotting was used to determine the protein level of ΔFosB and μ-opioid receptor proteins in the striatum and the midbrain of male offspring, respectively. Results showed that maternal morphine administration dramatically increased anxiety-like and morphine place preference behaviors in offspring. Also, ISO condition aggravated these behavioral outcomes. While, rearing in EE could attenuate anxiety and morphine conditioning in pups. At molecular levels, maternal morphine exposure and social isolation markedly increased both of ΔFosB and μ-opioid receptor proteins expression. However, rearing in the EE declined ΔFosB protein expression. Together, these findings help to elucidate long lasting impacts of early life morphine exposure and rearing environment on the behavioral and molecular profile of addicted individuals.
Collapse
Affiliation(s)
- Neda Yazdanfar
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Neuroscience, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Farnam
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Alireza Sarkaki
- The Persian Gulf Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
19
|
Radhakrishnan R, Grecco G, Stolze K, Atwood B, Jennings SG, Lien IZ, Saykin AJ, Sadhasivam S. Neuroimaging in infants with prenatal opioid exposure: Current evidence, recent developments and targets for future research. J Neuroradiol 2021; 48:112-120. [PMID: 33065196 PMCID: PMC7979441 DOI: 10.1016/j.neurad.2020.09.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/29/2022]
Abstract
Prenatal opioid exposure (POE) has shown to be a risk factor for adverse long-term cognitive and behavioral outcomes in offspring. However, the neural mechanisms of these outcomes remain poorly understood. While preclinical and human studies suggest that these outcomes may be due to opioid-mediated changes in the fetal and early postnatal brain, other maternal, social, and environmental factors are also shown to play a role. Recent neuroimaging studies reveal brain alterations in children with POE. Early neuroimaging and novel methodology could provide an in vivo mechanistic understanding of opioid mediated alterations in developing brain. However, this is an area of ongoing research. In this review we explore recent imaging developments in POE, with emphasis on the neonatal and infant brain, and highlight some of the challenges of imaging the developing brain in this population. We also highlight evidence from animal models and imaging in older children and youth to understand areas where future research may be targeted in infants with POE.
Collapse
Affiliation(s)
- Rupa Radhakrishnan
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| | - Gregory Grecco
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Brady Atwood
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Samuel G Jennings
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Izlin Z Lien
- Department of Pediatrics, Division of Neonatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | |
Collapse
|
20
|
Hocker AD, Morrison NR, Selby ML, Huxtable AG. Maternal Methadone Destabilizes Neonatal Breathing and Desensitizes Neonates to Opioid-Induced Respiratory Frequency Depression. Front Physiol 2021; 12:604593. [PMID: 33716765 PMCID: PMC7946987 DOI: 10.3389/fphys.2021.604593] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 02/08/2021] [Indexed: 11/28/2022] Open
Abstract
Pregnant women and developing infants are understudied populations in the opioid crisis, despite the rise in opioid use during pregnancy. Maternal opioid use results in diverse negative outcomes for the fetus/newborn, including death; however, the effects of perinatal (maternal and neonatal) opioids on developing respiratory circuitry are not well understood. Given the profound depressive effects of opioids on central respiratory networks controlling breathing, we tested the hypothesis that perinatal opioid exposure impairs respiratory neural circuitry, creating breathing instability. Our data demonstrate maternal opioids increase apneas and destabilize neonatal breathing. Maternal opioids also blunted opioid-induced respiratory frequency depression acutely in neonates; a unique finding since adult respiratory circuity does not desensitize to opioids. This desensitization normalized rapidly between postnatal days 1 and 2 (P1 and P2), the same age quantal slowing emerged in respiratory rhythm. These data suggest significant reorganization of respiratory rhythm generating circuits at P1-2, the same time as the preBötzinger Complex (key site of respiratory rhythm generation) becomes the dominant respiratory rhythm generator. Thus, these studies provide critical insight relevant to the normal developmental trajectory of respiratory circuits and suggest changes to mutual coupling between respiratory oscillators, while also highlighting how maternal opioids alter these developing circuits. In conclusion, the results presented demonstrate neurorespiratory disruption by maternal opioids and blunted opioid-induced respiratory frequency depression with neonatal opioids, which will be important for understanding and treating the increasing population of neonates exposed to gestational opioids.
Collapse
|
21
|
Newville J, Maxwell JR, Kitase Y, Robinson S, Jantzie LL. Perinatal Opioid Exposure Primes the Peripheral Immune System Toward Hyperreactivity. Front Pediatr 2020; 8:272. [PMID: 32670993 PMCID: PMC7332770 DOI: 10.3389/fped.2020.00272] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/29/2020] [Indexed: 11/29/2022] Open
Abstract
The increased incidence of opioid use during pregnancy warrants investigation to reveal the impact of opioid exposure on the developing fetus. Exposure during critical periods of development could have enduring consequences for affected individuals. Particularly, evidence is mounting that developmental injury can result in immune priming, whereby subsequent immune activation elicits an exaggerated immune response. This maladaptive hypersensitivity to immune challenge perpetuates dysregulated inflammatory signaling and poor health outcomes. Utilizing an established preclinical rat model of perinatal methadone exposure, we sought to investigate the consequences of developmental opioid exposure on in vitro activation of peripheral blood mononuclear cells (PBMCs). We hypothesize that PBMCs from methadone-exposed rats would exhibit abnormal chemokine and cytokine expression at baseline, with exaggerated chemokine and cytokine production following immune stimulation compared to saline-exposed controls. On postnatal day (P) 7, pup PMBCs were isolated and cultured, pooling three pups per n. Following 3 and 24 h, the supernatant from cultured PMBCs was collected and assessed for inflammatory cytokine and chemokine expression at baseline or lipopolysaccharide (LPS) stimulation using multiplex electrochemiluminescence. Following 3 and 24 h, baseline production of proinflammatory chemokine and cytokine levels were significantly increased in methadone PBMCs (p < 0.0001). Stimulation with LPS for 3 h resulted in increased tumor necrosis factor (TNF-α) and C-X-C motif chemokine ligand 1 (CXCL1) expression by 3.5-fold in PBMCs from methadone-exposed PBMCs compared to PBMCs from saline-exposed controls (p < 0.0001). Peripheral blood mononuclear cell hyperreactivity was still apparent at 24 h of LPS stimulation, evidenced by significantly increased TNF-α, CXCL1, interleukin 6 (IL-6), and IL-10 production by methadone PMBCs compared to saline control PBMCs (p < 0.0001). Together, we provide evidence of increased production of proinflammatory molecules from methadone PBMCs at baseline, in addition to sustained hyperreactivity relative to saline-exposed controls. Exaggerated peripheral immune responses exacerbate inflammatory signaling, with subsequent consequences on many organ systems throughout the body, such as the developing nervous system. Enhanced understanding of these inflammatory mechanisms will allow for appropriate therapeutic development for infants who were exposed to opioids during development. Furthermore, these data highlight the utility of this in vitro PBMC assay technique for future biomarker development to guide specific treatment for patients exposed to opioids during gestation.
Collapse
Affiliation(s)
- Jessie Newville
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Jessie R. Maxwell
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
- Departments of Pediatrics, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Yuma Kitase
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shenandoah Robinson
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lauren L. Jantzie
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Kennedy Krieger Institute, Baltimore, MD, United States
| |
Collapse
|
22
|
Brain microstructural changes support cognitive deficits in HIV uninfected children born to HIV infected mothers. Brain Behav Immun Health 2020; 2:100039. [PMID: 34589830 PMCID: PMC8474176 DOI: 10.1016/j.bbih.2020.100039] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/20/2020] [Accepted: 01/24/2020] [Indexed: 12/22/2022] Open
Abstract
Introduction Antiretroviral therapy (ART) is considered the most effective way to prevent perinatal transmission of human immunodeficiency virus (HIV). However, there is little knowledge about the effect of ART on the brain of HIV uninfected children born to HIV infected mothers (HUC). The current study evaluated the brain's microstructural integrity, and cognitive function in HUC compared to healthy children born to normal mothers (CHNM) and HIV infected children born to HIV infected mothers (HIC) to investigate the effect of in-utero exposure of ART on cerebral gray and white matter. Materials and methods Forty nine HIC, 12 HUC and 18 CHNM underwent neuropsychological (NP) assessment and a brain MRI. Diffusion tensor imaging (DTI) data was used to generate fractional anisotropy (FA) and mean diffusivity (MD) maps. Voxel wise comparison for FA and MD was performed between three groups using an analysis of covariance (ANCOVA) including age and sex as covariates, and correction for multiple comparisons (false discovery rate (FDR), p < 0.05 with minimum extended cluster size, 150 voxels). NP test scores were also compared between three groups using ANOVA with Post Hoc Bonferroni multiple comparison corrections (p < 0.05). Significantly changed FA and MD values in different brain regions in HIC and HUC compared to CHNM were used for correlation analysis with NP test scores using Pearson's correlation. Results HIC and HUC groups showed significantly decreased NP test scores in various domain compared to CHNM. Significantly lower NP test scores was observed in HIC than those of HUC. HIC showed decreased FA and increased MD in multiple brain sites as compared to both CHNM and HUC. Decreased FA along with both increased and decreased MD in different brain regions was present in HUC compared to CHNM. Both positive and negative correlation of altered FA and MD values from different brain regions in HIC and HUC with NP test scores was observed. Conclusion The presence of brain tissue changes and neurocognitive function deficit in absence of HIV infection in HUC indicates that ART may have a detrimental impact on the developing brain. The findings of the current study underscore the need for screening of ART exposed children for neurodevelopment and cognitive abnormalities at an early stage and call for access to early interventions, and nutritional and care programs.
Collapse
|
23
|
Ward P, Moss HG, Brown TR, Kalivas P, Jenkins DD. N-acetylcysteine mitigates acute opioid withdrawal behaviors and CNS oxidative stress in neonatal rats. Pediatr Res 2020; 88:77-84. [PMID: 31935745 PMCID: PMC7326708 DOI: 10.1038/s41390-019-0728-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 11/04/2019] [Accepted: 11/21/2019] [Indexed: 01/12/2023]
Abstract
BACKGROUND Neonatal abstinence syndrome (NAS) is a significant problem. Opioid withdrawal induces oxidative stress and disrupts glutamate and glutathione homeostasis. We hypothesized that N-acetylcysteine (NAC) administered during acute opioid withdrawal in neonatal rats would decrease withdrawal behaviors and normalize CNS glutathione and glutamate. METHODS Osmotic minipumps with methadone (opioid dependent, OD) and saline (Sham) were implanted into Sprague Dawley dams 7 days prior to delivery. Pups were randomized to receive either naloxone plus saline or NAC (50-100 mg/kg), administered on postnatal day (PND) 7. We performed MR spectroscopy on PND6-7 before, 30 min, and 120 min after withdrawal. On PND7, we assessed withdrawal behaviors for 90 min after naloxone administration and summed scores during peak withdrawal period. RESULTS Mean summed behavioral scores were significantly different between groups (χ2 (2) = 10.49, p = 0.005) but not different between NAC/NAL/OD and Sham (p = 0.14): SAL/NAL/OD = 17.2 ± 4.2 (n = 10); NAC/NAL/OD = 11.3 ± 5.6 (n = 9); Sham = 6.5 ± 0.6 (n = 4). SAL/NAL/OD pups had decreased glutathione at 120 min (p = 0.01), while NAC/NAL/OD pups maintained pre-withdrawal glutathione (p = 0.26). CONCLUSION In antenatal OD, NAC maintains CNS glutathione and mitigates acute opioid withdrawal in neonatal rats. This is the first study to demonstrate acute opioid withdrawal neurochemical changes in vivo in neonatal OD. NAC is a potential novel treatment for NAS.
Collapse
Affiliation(s)
- Price Ward
- 0000 0001 2189 3475grid.259828.cDepartment of Pediatrics, Medical University of South Carolina, Charleston, SC USA
| | - Hunter G. Moss
- 0000 0001 2189 3475grid.259828.cDepartment of Neuroscience, Medical University of South Carolina, Charleston, SC USA
| | - Truman R. Brown
- 0000 0001 2189 3475grid.259828.cDepartment of Radiology, Medical University of South Carolina, Charleston, SC USA
| | - Peter Kalivas
- 0000 0001 2189 3475grid.259828.cDepartment of Neuroscience, Medical University of South Carolina, Charleston, SC USA
| | - Dorothea D. Jenkins
- 0000 0001 2189 3475grid.259828.cDepartment of Pediatrics, Medical University of South Carolina, Charleston, SC USA
| |
Collapse
|
24
|
Caritis SN, Panigrahy A. Opioids affect the fetal brain: reframing the detoxification debate. Am J Obstet Gynecol 2019; 221:602-608. [PMID: 31323217 DOI: 10.1016/j.ajog.2019.07.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/26/2019] [Accepted: 07/12/2019] [Indexed: 10/26/2022]
Abstract
Medication-assisted treatment is recommended for individuals with an opioid use disorder, including pregnant women. Medication-assisted treatment during pregnancy provides benefits to the mother and fetus, including better pregnancy outcomes, reduced illicit drug use, and improved prenatal care. An alternative approach, medically supervised withdrawal (detoxification), has, in recent reports, demonstrated a low risk of fetal death and low rates of relapse and neonatal abstinence syndrome. The rates of relapse and neonatal abstinence syndrome are questioned by many who view medically supervised withdrawal as unacceptable based on the concern for the potential adverse consequences of relapse to mother and baby. The impact of opioids on the fetal brain have not been integrated into this debate. Studies in animals and human brain tissues demonstrate opioid receptors in neurons, astroglia, and oligodendrocytes. Age-specific normative data from infants, children, and adults have facilitated investigation of the impact of opioids on the human brain in vivo. Collectively, these studies in animals, human neural tissue, adult brains, and the brains of children and newborns demonstrate that opioids adversely affect the human brain, primarily the developing oligodendrocyte and the processes of myelinization (white matter microstructure), connectivity between parts of the brain, and the size of multiple brain regions, including the basal ganglia, thalamus, and cerebellar white matter. These in vivo studies across the human lifespan suggest vulnerability of specific fronto-temporal-limbic and frontal-subcortical (basal ganglia and cerebellum) pathways that are also likely vulnerable in the human fetal brain. The long-term impact of these reproducible changes in the fetal brain in vivo is unclear, but the possibility of lasting injury has been suggested. In light of the recent data on medically supervised withdrawal and the emerging evidence suggesting adverse effects of opioids on the developing fetal brain, a new paradigm of care is needed that includes the preferred option of medication-assisted treatment but also the option of medically supervised opioid withdrawal for a select group of women. Both these treatment options should offer mental health and social services support throughout pregnancy. More research on both opioid exposure on the developing human brain and the impact of medically supervised withdrawal is required to identify appropriate candidates, optimal dose reduction regimens, and gestational age timing for initiating medically supervised withdrawal.
Collapse
|
25
|
Merhar SL, Parikh NA, Braimah A, Poindexter BB, Tkach J, Kline-Fath B. White Matter Injury and Structural Anomalies in Infants with Prenatal Opioid Exposure. AJNR Am J Neuroradiol 2019; 40:2161-2165. [PMID: 31624119 DOI: 10.3174/ajnr.a6282] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/03/2019] [Indexed: 01/24/2023]
Abstract
Previous studies have not found structural injury or brain malformations in infants and children with prenatal opioid exposure. As part of an ongoing study evaluating neuroimaging in infants with prenatal opioid exposure, we reviewed structural brain MR imaging in 20 term infants with prenatal opioid exposure and 20 term controls at 4-8 weeks of age. We found that 8 of the 20 opioid-exposed infants had punctate white matter lesions or white matter signal abnormality on structural MR imaging, and 2 of the opioid-exposed infants had a septopreoptic fusion anomaly. No controls had white matter injury or structural malformations. Our findings underscore the importance of clinical neurodevelopmental follow-up and the need for more comprehensive imaging and long-term outcomes research following prenatal opioid exposure.
Collapse
Affiliation(s)
- S L Merhar
- From the Perinatal Institute, Division of Neonatology (S.L.M., N.A.P., B.B.P.)
- Department of Pediatrics (S.L.M., N.A.P., B.B.P.), University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - N A Parikh
- From the Perinatal Institute, Division of Neonatology (S.L.M., N.A.P., B.B.P.)
- Department of Pediatrics (S.L.M., N.A.P., B.B.P.), University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - A Braimah
- Pediatric Neuroimaging Research Consortium (A.B.)
| | - B B Poindexter
- From the Perinatal Institute, Division of Neonatology (S.L.M., N.A.P., B.B.P.)
- Department of Pediatrics (S.L.M., N.A.P., B.B.P.), University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - J Tkach
- Department of Radiology (J.T., B.K.-F.), Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - B Kline-Fath
- Department of Radiology (J.T., B.K.-F.), Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
26
|
The imperative of clinical and molecular research on neonatal opioid withdrawal syndrome. Mol Psychiatry 2019; 24:1568-1571. [PMID: 31506603 PMCID: PMC7983191 DOI: 10.1038/s41380-019-0522-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 08/02/2019] [Indexed: 11/09/2022]
|
27
|
Lee SJ, Woodward LJ, Henderson JMT. Educational achievement at age 9.5 years of children born to mothers maintained on methadone during pregnancy. PLoS One 2019; 14:e0223685. [PMID: 31600325 PMCID: PMC6786534 DOI: 10.1371/journal.pone.0223685] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/25/2019] [Indexed: 02/08/2023] Open
Abstract
Recent research shows that preschool children born to opioid-dependent mothers are at increased risk for cognitive, psychomotor, attention, and social-emotional adjustment problems. But very little is known about their school-age functioning, particularly their educational achievement. This analysis examined the educational outcomes of a regional cohort of 100 prenatally methadone-exposed children who were prospectively studied from birth to age 9.5 years alongside a comparison group of 110 randomly identified non-exposed children born between 2003 and 2008. At age 9.5, as part of a comprehensive neurodevelopmental evaluation, children's teachers rated their achievement across the school curriculum, and children completed the Woodcock Johnson-III Tests of Achievement (WJ-III). Detailed information about the birth mother's social background, pregnancy substance use, and mental health was also collected during pregnancy/at term. Infant clinical data were collected after birth. Methadone-exposed children performed less well than non-exposed children across seven school curriculum areas rated by teachers (ps ≤.001), performed less well than non-exposed children on all reading and mathematics subtests of the WJ-III, and had higher rates of any educational delay on the WJ-III (57% vs. 15%), OR = 7.47 (3.71-15.02). Results were similar when children with severe intellectual impairment were excluded. After adjusting for confounding factors, methadone-exposed children had increased odds of educational delay, but this was only marginally significant (OR = 3.62, [1.01-13.01], p = .049). Maternal educational attainment level (OR = 0.69, [0.50-0.89], p = .006), and maternal benzodiazepine use during pregnancy (OR = 2.70 [1.03-7.12], p = .044) were also associated with later educational risk. Findings suggest that children born to opioid-dependent women enrolled in methadone maintenance are at high risk of educational delay by age 9.5 years. Children's academic difficulties appeared to reflect the effects of both adverse prenatal exposures and postnatal social risk.
Collapse
Affiliation(s)
- Samantha J. Lee
- School of Psychology, Speech and Hearing, University of Canterbury, Christchurch, New Zealand
- * E-mail: (SJL); (JMTH)
| | - Lianne J. Woodward
- School of Health Sciences, University of Canterbury, Christchurch, New Zealand
| | - Jacqueline M. T. Henderson
- School of Psychology, Speech and Hearing, University of Canterbury, Christchurch, New Zealand
- * E-mail: (SJL); (JMTH)
| |
Collapse
|
28
|
Lambert JE, Peeler CE. Visual and oculomotor outcomes in children with prenatal opioid exposure. Curr Opin Ophthalmol 2019; 30:449-453. [PMID: 31449086 DOI: 10.1097/icu.0000000000000604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW To summarize the visual and oculomotor outcomes in children with prenatal opioid exposure and review the effects of opioids on the developing central nervous system. RECENT FINDINGS Animal models and imaging studies in children suggest that prenatal opioid exposure may affect neuronal survival and result in delayed maturation of white matter tracts and decreased volumes in certain brain areas. Visual evoked potential testing in children demonstrates delayed maturation of the afferent visual system in opioid-exposed groups compared with controls, though 'catch-up' development is seen with longitudinal follow-up. Strabismus and nystagmus are also more common in exposed children, and these findings appear to persist. SUMMARY As rates of opioid dependence and prenatal opioid exposure continue to increase, it is important to evaluate the short-term and long-term effects of opioids on the developing visual system. An understanding of these risks is important when counseling the parents or guardians of opioid-exposed children, though larger studies with more long-term follow-up will improve our prognostic abilities.
Collapse
Affiliation(s)
| | - Crandall E Peeler
- Department of Ophthalmology.,Department of Neurology, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
29
|
Morie KP, Crowley MJ, Mayes LC, Potenza MN. Prenatal drug exposure from infancy through emerging adulthood: Results from neuroimaging. Drug Alcohol Depend 2019; 198:39-53. [PMID: 30878766 PMCID: PMC6688747 DOI: 10.1016/j.drugalcdep.2019.01.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 01/15/2019] [Accepted: 01/15/2019] [Indexed: 11/28/2022]
Abstract
Prenatal drug exposure may have important repercussions across the lifespan for cognition and behavior. While alcohol is a recognized teratogen, the influences of other substances may also be substantial. The neural underpinnings of the influences of prenatal drug exposure have been examined using longitudinal approaches and multiple imaging techniques. Here we review the existing literature on the neural correlates of prenatal drug exposure. We focused the review on studies that have employed functional neuroimaging and electroencephalography and on substances other than alcohol. We also framed the review through the lens of four developmental life stages (infancy, childhood, adolescence and emerging adulthood). We included papers that have examined any drug use, including tobacco, opiates, cocaine, marijuana, methamphetamines, or polysubstance use. Data suggest that prenatal drug exposure has long-lasting, deleterious influences on cognition and reward processing in infancy and childhood that persist into adolescence and emerging adulthood and may underlie some behavioral tendencies, such as increased externalizing and risk-taking behaviors, seen in these groups.
Collapse
Affiliation(s)
- Kristen P. Morie
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA,Child Study Center, Yale University School of Medicine, New Haven, CT, 06510, USA,Corresponding author at: Department of Psychiatry, Yale University School of Medicine, 300 George St., #901, New Haven, CT, 06510, USA. (K.P. Morie)
| | - Michael J. Crowley
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA,Child Study Center, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Linda C. Mayes
- Child Study Center, Yale University School of Medicine, New Haven, CT, 06510, USA,Department of Pediatrics, Yale University School of Medicine, New Haven, CT, 06510, USA,Department of Psychology, Yale University, New Haven, CT, 06511, USA
| | - Marc N. Potenza
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA,Child Study Center, Yale University School of Medicine, New Haven, CT, 06510, USA,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA,Connecticut Mental Health Center, New Haven, CT, 06519, USA,Connecticut Council on Problem Gambling, Wethersfield, CT, 06109, USA
| |
Collapse
|
30
|
Walhovd KB, Howell GR, Ritchie SJ, Staff RT, Cotman CW. What are the earlier life contributions to reserve and resilience? Neurobiol Aging 2019; 83:135-139. [PMID: 31307838 DOI: 10.1016/j.neurobiolaging.2019.04.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 04/22/2019] [Indexed: 12/30/2022]
Abstract
The brain's structures and functions arise from a combination of developmental processes and interaction with environmental experiences, beginning in utero and continuing throughout the lifespan. Broadly, the process that we think of as "successful aging" likely has its foundation in early life and is continuously shaped as life experiences are programmed into the brain in response to a changing environment. Thus, individual lifestyle choices and interventions aimed at increasing cognitive reserve and resilience could change the course of cognitive aging. To determine the relative efficacy of these approaches, we will need to understand how the timing of these interventions (e.g., age, duration, frequency) influences cognitive capacity through the lifespan. Although analysis of age-related changes in cognitive function reveals a general decline at the population level, it has become clear that there is great individual variance in the extent to which cognitive function changes with advanced age. The factors responsible for the individual differences in cognitive decline are unclear, but uncovering them with new analytical tools, epigenetic approaches, and subpopulation studies will provide a roadmap toward enhancing reserve and resilience in the population at large and preserving cognitive function in a greater number of aging individuals.
Collapse
Affiliation(s)
- Kristine B Walhovd
- Department of Psychology, Center for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway
| | | | - Stuart J Ritchie
- Social, Genetic and Developmental Psychiatry Centre, King's College London, London, UK
| | - Roger T Staff
- Aberdeen Royal Infirmary, NHS Grampian, Scotland, UK
| | - Carl W Cotman
- Institute for Brain Aging and Dementia, University of California, Irvine, CA, USA.
| |
Collapse
|
31
|
Gao W, Grewen K, Knickmeyer RC, Qiu A, Salzwedel A, Lin W, Gilmore JH. A review on neuroimaging studies of genetic and environmental influences on early brain development. Neuroimage 2019; 185:802-812. [PMID: 29673965 PMCID: PMC6191379 DOI: 10.1016/j.neuroimage.2018.04.032] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 04/11/2018] [Accepted: 04/13/2018] [Indexed: 12/11/2022] Open
Abstract
The past decades witnessed a surge of interest in neuroimaging study of normal and abnormal early brain development. Structural and functional studies of normal early brain development revealed massive structural maturation as well as sequential, coordinated, and hierarchical emergence of functional networks during the infancy period, providing a great foundation for the investigation of abnormal early brain development mechanisms. Indeed, studies of altered brain development associated with either genetic or environmental risks emerged and thrived. In this paper, we will review selected studies of genetic and environmental risks that have been relatively more extensively investigated-familial risks, candidate risk genes, and genome-wide association studies (GWAS) on the genetic side; maternal mood disorders and prenatal drug exposures on the environmental side. Emerging studies on environment-gene interactions will also be reviewed. Our goal was not to perform an exhaustive review of all studies in the field but to leverage some representative ones to summarize the current state, point out potential limitations, and elicit discussions on important future directions.
Collapse
Affiliation(s)
- Wei Gao
- Biomedical Imaging Research Institute (BIRI), Department of Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, CA, United States; Department of Medicine, University of California, Los Angeles, CA, United States.
| | - Karen Grewen
- Department of Psychiatry, Neurobiology, and Psychology, University of North Carolina Chapel Hill, Chapel Hill, NC, United States
| | - Rebecca C Knickmeyer
- Department of Psychiatry, University of North Carolina at Chapel Hill, N.C, United States
| | - Anqi Qiu
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | - Andrew Salzwedel
- Biomedical Imaging Research Institute (BIRI), Department of Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, CA, United States
| | - Weili Lin
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, NC, United States
| | - John H Gilmore
- Department of Psychiatry, University of North Carolina at Chapel Hill, N.C, United States
| |
Collapse
|
32
|
Nygaard E, Slinning K, Moe V, Due-Tønnessen P, Fjell A, Walhovd KB. Neuroanatomical characteristics of youths with prenatal opioid and poly-drug exposure. Neurotoxicol Teratol 2018; 68:13-26. [PMID: 29679636 DOI: 10.1016/j.ntt.2018.04.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 03/21/2018] [Accepted: 04/16/2018] [Indexed: 12/12/2022]
Abstract
Neuroanatomical and cognitive differences have been documented during childhood between children with prenatal opioid- and poly-drug exposure and controls in small samples. We investigated whether these differences persisted in larger samples of youth at older ages. Quantitative MRI and cognitive data were compared between 38 youths in the risk group and 44 youths in the non-exposed group (aged 17 to 22 years) who had been followed prospectively since birth. Most drug-exposed youths (84%) moved to permanent foster or adoptive homes before one year of age. The drug-exposed group displayed smaller neuroanatomical volumes (0.70 SD difference in total brain volume, p = 0.001), smaller cortical surface areas and thinner cortices than the comparison group. The birth weight accounted for some of the intergroup differences. Neuroanatomical characteristics partially mediated group differences in cognitive function. The present study cannot differentiate between causal factors but indicates persistent neurocognitive differences associated with prenatal opioid or poly-drug exposure.
Collapse
Affiliation(s)
- Egil Nygaard
- Department of Psychology, University of Oslo, Postbox 1094 Blindern, 0317 Oslo, Norway; Center for Child and Adolescent Mental Health, Eastern and Southern Norway (RBUP), Postbox 4623 Nydalen, 0405 Oslo, Norway.
| | - Kari Slinning
- Department of Psychology, University of Oslo, Postbox 1094 Blindern, 0317 Oslo, Norway; Center for Child and Adolescent Mental Health, Eastern and Southern Norway (RBUP), Postbox 4623 Nydalen, 0405 Oslo, Norway.
| | - Vibeke Moe
- Department of Psychology, University of Oslo, Postbox 1094 Blindern, 0317 Oslo, Norway; Center for Child and Adolescent Mental Health, Eastern and Southern Norway (RBUP), Postbox 4623 Nydalen, 0405 Oslo, Norway.
| | - Paulina Due-Tønnessen
- Department of Psychology, University of Oslo, Postbox 1094 Blindern, 0317 Oslo, Norway; Department of Radiology, Rikshospitalet University Hospital, Oslo, Norway.
| | - Anders Fjell
- Department of Psychology, University of Oslo, Postbox 1094 Blindern, 0317 Oslo, Norway.
| | - Kristine B Walhovd
- Department of Psychology, University of Oslo, Postbox 1094 Blindern, 0317 Oslo, Norway.
| |
Collapse
|
33
|
Sirnes E, Griffiths ST, Aukland SM, Eide GE, Elgen IB, Gundersen H. Functional MRI in prenatally opioid-exposed children during a working memory-selective attention task. Neurotoxicol Teratol 2018; 66:46-54. [PMID: 29408607 DOI: 10.1016/j.ntt.2018.01.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/11/2018] [Accepted: 01/31/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Opioid induced cerebral changes may contribute to neuropsychological difficulties, like attention problems, frequently reported in prenatally opioid-exposed children. Reduced regional brain volumes have been shown after prenatal opioid exposure, but no study to date has explored the possible impact of prenatal opioids on brain activation patterns. MATERIALS AND METHODS A hospital-based sample of prenatally opioid-exposed school-aged children (n = 11) and unexposed controls (n = 12) underwent functional magnetic resonance imaging (fMRI) during a combined working memory-selective attention task. Within-group- and between-group analyses of blood-oxygen-level-dependent (BOLD) activation were performed using the SPM12 software package and group differences in task performance were analyzed using Cox proportional hazards modeling. RESULTS Overall, similar patterns of task related parietal and prefrontal BOLD activations were found in both groups. The opioid-exposed group showed impaired task performance, and during the most cognitive demanding versions of the working memory-selective attention task, increased activation in prefrontal cortical areas was found in the opioid-exposed group compared to controls. CONCLUSION Our findings suggest that prenatal opioids affect later brain function, visible through changes in BOLD activation patterns. However, results should be considered preliminary until replicated in larger samples better suited to control for potential confounding factors.
Collapse
Affiliation(s)
- Eivind Sirnes
- Department of Child and Adolescent Psychiatry, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway.
| | - Silja T Griffiths
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Stein Magnus Aukland
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - Geir Egil Eide
- Centre for Clinical Research, Haukeland University Hospital, Bergen, Norway; Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Irene B Elgen
- Department of Child and Adolescent Psychiatry, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Hilde Gundersen
- Department of Sport and Physical Education, Western Norway University of Applied Sciences, Bergen, Norway
| |
Collapse
|
34
|
Lind JN, Interrante JD, Ailes EC, Gilboa SM, Khan S, Frey MT, Dawson AL, Honein MA, Dowling NF, Razzaghi H, Creanga AA, Broussard CS. Maternal Use of Opioids During Pregnancy and Congenital Malformations: A Systematic Review. Pediatrics 2017; 139:e20164131. [PMID: 28562278 PMCID: PMC5561453 DOI: 10.1542/peds.2016-4131] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/07/2017] [Indexed: 11/24/2022] Open
Abstract
CONTEXT Opioid use and abuse have increased dramatically in recent years, particularly among women. OBJECTIVES We conducted a systematic review to evaluate the association between prenatal opioid use and congenital malformations. DATA SOURCES We searched Medline and Embase for studies published from 1946 to 2016 and reviewed reference lists to identify additional relevant studies. STUDY SELECTION We included studies that were full-text journal articles and reported the results of original epidemiologic research on prenatal opioid exposure and congenital malformations. We assessed study eligibility in multiple phases using a standardized, duplicate review process. DATA EXTRACTION Data on study characteristics, opioid exposure, timing of exposure during pregnancy, congenital malformations (collectively or as individual subtypes), length of follow-up, and main findings were extracted from eligible studies. RESULTS Of the 68 studies that met our inclusion criteria, 46 had an unexposed comparison group; of those, 30 performed statistical tests to measure associations between maternal opioid use during pregnancy and congenital malformations. Seventeen of these (10 of 12 case-control and 7 of 18 cohort studies) documented statistically significant positive associations. Among the case-control studies, associations with oral clefts and ventricular septal defects/atrial septal defects were the most frequently reported specific malformations. Among the cohort studies, clubfoot was the most frequently reported specific malformation. LIMITATIONS Variabilities in study design, poor study quality, and weaknesses with outcome and exposure measurement. CONCLUSIONS Uncertainty remains regarding the teratogenicity of opioids; a careful assessment of risks and benefits is warranted when considering opioid treatment for women of reproductive age.
Collapse
Affiliation(s)
- Jennifer N Lind
- Division of Congenital and Developmental Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia;
- US Public Health Service, Atlanta, Georgia
| | - Julia D Interrante
- Division of Congenital and Developmental Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee
| | - Elizabeth C Ailes
- Division of Congenital and Developmental Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Suzanne M Gilboa
- Division of Congenital and Developmental Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Sara Khan
- Division of Congenital and Developmental Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia
- Carter Consulting, Atlanta, Georgia; and
| | - Meghan T Frey
- Division of Congenital and Developmental Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - April L Dawson
- Division of Congenital and Developmental Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Margaret A Honein
- Division of Congenital and Developmental Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Nicole F Dowling
- Division of Congenital and Developmental Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Hilda Razzaghi
- Division of Congenital and Developmental Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia
- US Public Health Service, Atlanta, Georgia
| | - Andreea A Creanga
- Department of International Health and
- International Center for Maternal and Newborn Health, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Cheryl S Broussard
- Division of Congenital and Developmental Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
35
|
Morie KP, Yip SW, Zhai ZW, Xu J, Hamilton KR, Mayes LC, Potenza MN, Potenza MN. White-matter crossing-fiber microstructure in adolescents prenatally exposed to cocaine. Drug Alcohol Depend 2017; 174:23-29. [PMID: 28292689 PMCID: PMC5555052 DOI: 10.1016/j.drugalcdep.2017.01.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 01/05/2017] [Accepted: 01/07/2017] [Indexed: 11/26/2022]
Abstract
BACKGROUND Prenatal cocaine exposure (PCE) is associated with risk-taking behaviors, including increased initiation of substance use in adolescence. The neurobiological underpinnings of these behaviors in adolescents with PCE are not well understood. The goal of this study was to compare diffusion-weighted imaging data between adolescents with and without PCE using crossing-fiber models, which may provide more comprehensive estimates of white-matter microstructure within regions of multiple (e.g., primary and secondary) fiber orientations. METHODS Thirty-nine PCE individuals and 17 comparably aged prenatally non-drug-exposed (NDE) youths were recruited from a longitudinal cohort followed since birth. White matter was examined using tensor-derived and crossing-fiber models. Whole-brain investigations were performed, as were analyses on seven white-matter regions, which included the splenium, body and genu of the corpus callosum, bilateral cingulum, and the right and left superior longitudinal fasciculus (SLF). RESULTS Whole-brain analyses revealed no group differences. However, ROI analyses for anisotropy estimates derived from the crossing-fiber model revealed significant group differences for secondary fibers, with reduced anisotropy among PCE adolescents compared to prenatally non-exposed youth in the right cingulum and the left SLF, and increased anisotropy in the genu. CONCLUSIONS Our findings suggest that white-matter differences in PCE adolescents are subtle and localized primarily within secondary fiber orientations, perhaps arising from altered white-matter development.
Collapse
Affiliation(s)
- Kristen P. Morie
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA,Corresponding author: Kristen Morie,
| | - Sarah W. Yip
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA,National Center on Addictions and Substance Abuse, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Zu Wei Zhai
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Jiansong Xu
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Kristen R. Hamilton
- Department of Psychology, University of Maryland, College Park, MD, 20742 USA
| | - Linda C. Mayes
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA,Child Study Center, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Marc N. Potenza
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA,National Center on Addictions and Substance Abuse, Yale University School of Medicine, New Haven, CT, 06510, USA,Child Study Center, Yale University School of Medicine, New Haven, CT, 06510, USA,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA,Connecticut Mental Health Center, New Haven, CT, 06519, USA
| | - Marc N Potenza
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06510, USA; National Center on Addictions and Substance Abuse, Yale University School of Medicine, New Haven, CT, 06510, USA; Child Study Center, Yale University School of Medicine, New Haven, CT, 06510, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA; Connecticut Mental Health Center, New Haven, CT, 06519, USA
| |
Collapse
|
36
|
A Review of Neurobehavioral Challenges in Children Exposed Prenatally to Intrauterine Opioid. JOURNAL OF PEDIATRICS REVIEW 2017. [DOI: 10.5812/jpr.9234] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
37
|
Brain morphology in school-aged children with prenatal opioid exposure: A structural MRI study. Early Hum Dev 2017; 106-107:33-39. [PMID: 28187337 DOI: 10.1016/j.earlhumdev.2017.01.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 01/27/2017] [Accepted: 01/30/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Both animal and human studies have suggested that prenatal opioid exposure may be detrimental to the developing fetal brain. However, results are somewhat conflicting. Structural brain changes in children with prenatal opioid exposure have been reported in a few studies, and such changes may contribute to neuropsychological impairments observed in exposed children. AIM To investigate the association between prenatal opioid exposure and brain morphology in school-aged children. STUDY DESIGN A cross-sectional magnetic resonance imaging (MRI) study of prenatally opioid-exposed children and matched controls. SUBJECTS A hospital-based sample (n=16) of children aged 10-14years with prenatal exposure to opioids and 1:1 sex- and age-matched unexposed controls. OUTCOME MEASURES Automated brain volume measures obtained from T1-weighted MRI scans using FreeSurfer. RESULTS Volumes of the basal ganglia, thalamus, and cerebellar white matter were reduced in the opioid-exposed group, whereas there were no statistically significant differences in global brain measures (total brain, cerebral cortex, and cerebral white matter volumes). CONCLUSIONS In line with the limited findings reported in the literature to date, our study showed an association between prenatal opioid exposure and reduced regional brain volumes. Adverse effects of opioids on the developing fetal brain may explain this association. However, further research is needed to explore the causal nature and functional consequences of these findings.
Collapse
|
38
|
Vassoler FM, Oliver DJ, Wyse C, Blau A, Shtutman M, Turner JR, Byrnes EM. Transgenerational attenuation of opioid self-administration as a consequence of adolescent morphine exposure. Neuropharmacology 2017; 113:271-280. [PMID: 27729240 PMCID: PMC5248554 DOI: 10.1016/j.neuropharm.2016.10.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 09/29/2016] [Accepted: 10/07/2016] [Indexed: 01/13/2023]
Abstract
The United States is in the midst of an opiate epidemic, with abuse of prescription and illegal opioids increasing steadily over the past decade. While it is clear that there is a genetic component to opioid addiction, there is a significant portion of heritability that cannot be explained by genetics alone. The current study was designed to test the hypothesis that maternal exposure to opioids prior to pregnancy alters abuse liability in subsequent generations. Female adolescent Sprague Dawley rats were administered morphine at increasing doses (5-25 mg/kg, s.c.) or saline for 10 days (P30-39). During adulthood, animals were bred with drug-naïve colony males. Male and female adult offspring (F1 animals) were tested for morphine self-administration acquisition, progressive ratio, extinction, and reinstatement at three doses of morphine (0.25, 0.75, 1.25 mg/kg/infusion). Grandoffspring (F2 animals, from the maternal line) were also examined. Additionally, gene expression changes within the nucleus accumbens were examined with RNA deep sequencing (PacBio) and qPCR. There were dose- and sex-dependent effects on all phases of the self-administration paradigm that indicate decreased morphine reinforcement and attenuated relapse-like behavior. Additionally, genes related to synaptic plasticity, as well as myelin basic protein (MBP), were dysregulated. Some, but not all, effects persisted into the subsequent (F2) generation. The results demonstrate that even limited opioid exposure during adolescence can have lasting effects across multiple generations, which has implications for mechanisms of the transmission of drug abuse liability in humans.
Collapse
Affiliation(s)
- Fair M Vassoler
- Cummings School at Tufts University, 200 Westboro Road, North Grafton, MA 01536, USA.
| | - David J Oliver
- University of South Carolina College of Pharmacy, 773 Sumter St, Columbia, SC 29208, USA
| | - Cristina Wyse
- Cummings School at Tufts University, 200 Westboro Road, North Grafton, MA 01536, USA
| | - Ashley Blau
- Cummings School at Tufts University, 200 Westboro Road, North Grafton, MA 01536, USA
| | - Michael Shtutman
- University of South Carolina College of Pharmacy, 773 Sumter St, Columbia, SC 29208, USA
| | - Jill R Turner
- University of South Carolina College of Pharmacy, 773 Sumter St, Columbia, SC 29208, USA
| | - Elizabeth M Byrnes
- Cummings School at Tufts University, 200 Westboro Road, North Grafton, MA 01536, USA
| |
Collapse
|
39
|
Devlin LA, Lau T, Radmacher PG. Decreasing Total Medication Exposure and Length of Stay While Completing Withdrawal for Neonatal Abstinence Syndrome during the Neonatal Hospital Stay. Front Pediatr 2017; 5:216. [PMID: 29067285 PMCID: PMC5641300 DOI: 10.3389/fped.2017.00216] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 09/25/2017] [Indexed: 11/24/2022] Open
Abstract
INTRODUCTION Neonatal abstinence syndrome (NAS) is a rapidly growing public health concern that has considerably increased health-care utilization and health-care costs. In an effort to curtail costs, attempts have been made to complete withdrawal as an outpatient. Outpatient therapy has been shown to prolong exposure to medications, which may negatively impact neurodevelopmental and behavioral outcomes. We hypothesized that the implementation of a modified NAS protocol would decrease total drug exposure and length of stay while allowing for complete acute drug withdrawal during the neonatal hospital stay. METHODS Data were derived retrospectively from medical records of term (≥37 0/7) infants with NAS who were treated with pharmacologic therapy in the University of Louisville Hospital Neonatal Intensive Care Unit from 2005 to 2015. The pharmacologic protocol (SP1) for infants treated between 2005 and March 2014 (n = 146) dosed oral morphine every 4 h and utilized phenobarbital as adjuvant therapy. Protocol 2 (SP2) initiated after March 2014 (n = 44) dosed morphine every 3 h and used clonidine as adjuvant therapy. Charts were reviewed for demographic information and maternal drug history. Maternal and infant toxicology screens were recorded. The length of morphine therapy and need for adjuvant drug therapy were noted. Length of stay was derived from admission and discharge dates. RESULTS The length of morphine therapy was decreased by 8.5 days from 35 to 26.5 days (95% CI 4.5-12 days) for infants treated with SP2 vs. SP1 (p < 0.001). The need for adjuvant pharmacologic therapy was decreased by 24% in patients treated with SP2 vs. SP1 (p = 0.004). The length of stay was decreased by 9 days from 42 to 33 days (95% CI 5.1-13 days) for infants treated with SP2 vs. SP1 (p < 0.001). The decreased length of stay resulted in an average reduction of hospital charges by $27,090 per patient in adjusted 2015 US Dollars. CONCLUSION This study demonstrates that total drug exposure and length of stay can be reduced while successfully completing acute withdrawal during the neonatal hospital stay.
Collapse
Affiliation(s)
- Lori A Devlin
- Division of Neonatal Medicine, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States
| | - Timothy Lau
- Department of Educational and Counseling Psychology, University of Louisville, Louisville, KY, United States
| | - Paula G Radmacher
- Division of Neonatal Medicine, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States
| |
Collapse
|
40
|
Yepes-Calderon F, Lao Y, Fillard P, Nelson MD, Panigrahy A, Lepore N. Tractography in the clinics: Implementing a pipeline to characterize early brain development. NEUROIMAGE-CLINICAL 2016; 14:629-640. [PMID: 28348954 PMCID: PMC5357703 DOI: 10.1016/j.nicl.2016.12.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 02/06/2023]
Abstract
In imaging studies of neonates, particularly in the clinical setting, diffusion tensor imaging-based tractography is typically unreliable due to the use of fast acquisition protocols that yield low resolution and signal-to-noise ratio (SNR). These image acquisition protocols are implemented with the aim of reducing motion artifacts that may be produced by the movement of the neonate's head during the scanning session. Furthermore, axons are not yet fully myelinated in these subjects. As a result, the water molecules' movements are not as constrained as in older brains, making it even harder to define structure using diffusion profiles. Here, we introduce a post-processing method that overcomes the difficulties described above, allowing the determination of reliable tracts in newborns. We tested our method using neonatal data and successfully extracted some of the limbic, association and commissural fibers, all of which are typically difficult to obtain by direct tractography. Geometrical and diffusion based features of the tracts are then utilized to compare premature babies to term babies. Our results quantify the maturation of white matter fiber tracts in neonates. The proposed method enables consistent tractography in clinical datasets. The tractography is used to structural positioning purposes Geometrical features and diffusion variables in the tracts' paths are analyzed. The gestational age was predicted with regressions in term and preterm babies. The extracted features can be used as indexes of early neurodevelopment.
Collapse
Affiliation(s)
- Fernando Yepes-Calderon
- Childrens Hospital Los Angeles, Neurosurgery, 1300 Vermont Ave, Los Angeles, CA, USA; Universidad de Barcelona, Facultad de Medicina, Casanova 43, Barcelona, Spain
| | - Yi Lao
- Children Hospital Los Angeles, Radiology, 4650 Sunset Blvd, Los Angeles, CA, USA
| | - Pierre Fillard
- Parietal Research Team, INRIA Saclay le-de-France, Neurospin, France
| | - Marvin D Nelson
- Children Hospital Los Angeles, Radiology, 4650 Sunset Blvd, Los Angeles, CA, USA
| | - Ashok Panigrahy
- Children's Hospital of Pittsburgh, 4401 Penn Avenue Pittsburgh, Pittsburgh, PA, USA
| | - Natasha Lepore
- Children Hospital Los Angeles, Radiology, 4650 Sunset Blvd, Los Angeles, CA, USA
| |
Collapse
|
41
|
Gourévitch B, Cai J, Mellen N. Cellular and network-level adaptations to in utero methadone exposure along the ventral respiratory column in the neonate rat. Exp Neurol 2016; 287:S0014-4886(16)30063-2. [PMID: 27009496 DOI: 10.1016/j.expneurol.2016.03.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/17/2016] [Accepted: 03/18/2016] [Indexed: 11/15/2022]
Abstract
Neonatal abstinence syndrome (NAS) occurs in babies chronically exposed to opioids during pregnancy. NAS shares features with opioid withdrawal symptoms seen in adults, including autonomic dysregulation. Here, the effect of low-dose in utero methadone (MTD) exposure on respiration-modulated networks along the ventral respiratory column (VRC) in ventrolateral medulla was investigated in the neonate Sprague-Dawley rat. MTD was administered via drinking water (3mg/kg/day in drinking water of the mother E7-E21). Lower expression levels of myelin-associated proteins phosphorylated axonal neurofilament subunit H (pNFH), 2',3'-Cyclicnucleotide 3'-phosphodiesterase (CNPase) and myelin basic protein (MBP), in MTD-exposed pups compared to controls at P3, P6 and P10 indicated MTD transport across the placenta. We investigated whether in utero MTD exposure led to network-level excitability changes consistent with tolerance, and also probed for changes in endogenous opioid modulation of respiratory networks. To this end, high-speed (45.5Hz) optical recordings of respiratory network activity in control and MTD-exposed neonate (P0-P2) pups before and during administration of the μ-opioid receptor antagonist naloxone (NAL; 10μM) were carried out. Spike rate was estimated from optical traces via deconvolution, and coupling between all neuron pairs in recorded networks was quantified using the normalized transfer entropy (NTE). Recordings of local networks along the VRC, together with recordings of respiratory output from ventral root C1 did not reveal changes in respiratory activity at the system level, but cellular and network changes in MTD-exposed pups were consistent with the development of opioid tolerance. MTD-exposed pups were found to have i. higher neuronal firing rates; ii. higher covariance between neuronal activity and motor output; iii. more bidirectionally and unidirectionally coupled neurons, and fewer uncoupled neurons; iv. stronger coupling and shorter integration times between network constituents. The μ-opioid receptor antagonist NAL did not alter system-level function. The correlation between the activity of neurons caudal to -400μm and motor output was significantly reduced in control animals following NAL. In both control and MTD-exposed pups, the relative number of neurons whose correlation with motor output increased following NAL followed a rostrocaudal gradient along the VRC, with fewer neurons caudally, and more neurons rostrally. The up-regulation of coupling strength, firing rate and coefficient of variation between neurons and motor output following in utero opioid exposure suggests that these networks may contribute to NAS in infants born to opioid-dependent mothers.
Collapse
Affiliation(s)
- Boris Gourévitch
- NeuroPSI, UMR CNRS 8195, Bâtiment 446, 91405 Orsay cedex, France; Université Paris-Sud, Bâtiment 446, 91405 Orsay cedex, France
| | - Jun Cai
- Kosair Children's Hospital Research Institute, University of Louisville, Louisville, KY 40206, USA
| | - Nicholas Mellen
- Kosair Children's Hospital Research Institute, University of Louisville, Louisville, KY 40206, USA.
| |
Collapse
|
42
|
Methadone-induced Damage to White Matter Integrity in Methadone Maintenance Patients: A Longitudinal Self-control DTI Study. Sci Rep 2016; 6:19662. [PMID: 26794650 PMCID: PMC4726264 DOI: 10.1038/srep19662] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 12/10/2015] [Indexed: 11/22/2022] Open
Abstract
Methadone maintenance treatment (MMT) can induce impairments in brain function and structure, despite its clinical effectiveness. However, the effect of chronic MMT on brain white matter (WM) is not fully known. Thirty-three MMT patients underwent diffusion tensor imaging (DTI) twice – at the start of the study (Scan1) and one year later (Scan2). Tract-based spatial statistics were used to investigate changes in fractional anisotropy (FA), axial diffusivity (AD) and radial diffusivity (RD) between the two scans. The correlations between DTI indices and methadone consumption and neuropsychological status were analysed. We found significantly decreased FA, decreased AD and increased RD in Scan2 in extensive WM regions; overlapping regions were found in the left posterior limb and the retrolenticular part of internal capsule, superior and posterior corona radiata, bilateral external capsule and the right superior longitudinal fasciculus. In addition, the change of FA in the overlapping regions was positively correlated with the accumulated dosage of methadone use, the RD value in Scan2 and non-planning impulsiveness (NPI) measured at follow-up. The results suggest that methadone has damaging effects on WM integrity. The dose-dependent pattern and characteristics of the impairment may suggest new strategies for MMT.
Collapse
|
43
|
Nygaard E, Moe V, Slinning K, Walhovd KB. Longitudinal cognitive development of children born to mothers with opioid and polysubstance use. Pediatr Res 2015; 78:330-5. [PMID: 25978800 PMCID: PMC4539602 DOI: 10.1038/pr.2015.95] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 02/18/2015] [Indexed: 01/07/2023]
Abstract
BACKGROUND Previous studies indicate an increased risk for neuropsychological difficulties in young children prenatally exposed to opioids and polysubstances, but longitudinal information is scarce. The present longitudinal study investigated whether these waned, persisted, or increased over time. METHODS The cognitive functioning of 72 children with prenatal opioid and polysubstance exposure and 58 children without any established prenatal risk was assessed at 1, 2, 3, 4½, and 8½ y. RESULTS The exposed boys had significantly and stably lower levels of cognitive functioning than the control group, whereas there were increasing differences over time for the girls. The exposed group had significantly lower IQ scores than the control group on Wechsler Intelligence Scale for Children--Revised at 8½ y after controlling for earlier cognitive abilities, and for children who were permanently placed in adoptive/foster homes before 1 y of age and whose mothers used heroin as their main drug during pregnancy (B = 17.04, 95% CI 8.69-25.38, P < 0.001). CONCLUSION While effects of prenatal substance exposure cannot be isolated, group effects on cognition rather increased than waned over time, even in adoptive/foster children with minimal postnatal risk.
Collapse
Affiliation(s)
- Egil Nygaard
- Research Group for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo, Norway
- Center for Child and Adolescent Mental Health, Eastern and Southern Norway (RBUP), Oslo, Norway
| | - Vibeke Moe
- Center for Child and Adolescent Mental Health, Eastern and Southern Norway (RBUP), Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Kari Slinning
- Center for Child and Adolescent Mental Health, Eastern and Southern Norway (RBUP), Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Kristine B. Walhovd
- Research Group for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo, Norway
- Department of Physical Medicine and Rehabilition, Unit of Neuropsychology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
44
|
Isokuortti H, Iverson GL, Kataja A, Brander A, Öhman J, Luoto TM. Who Gets Head Trauma or Recruited in Mild Traumatic Brain Injury Research? J Neurotrauma 2015; 33:232-41. [PMID: 26054639 DOI: 10.1089/neu.2015.3888] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mild traumatic brain injury (mTBI) is a public health problem. Outcome from mTBI is heterogeneous in part due to pre-injury individual differences that typically are not well described or understood. Pre-injury health characteristics of all consecutive patients (n=3023) who underwent head computed tomography due to acute head trauma in the emergency department of Tampere University Hospital, Finland, between August 2010 and July 2012 were examined. Patients were screened to obtain a sample of working age adults with no pre-injury medical or mental health problems who had sustained a "pure" mTBI. Of all patients screened, 1990 (65.8%) fulfilled the mTBI criteria, 257 (8.5%) had a more severe TBI, and 776 (25.7%) had a head trauma without obvious signs of brain injury. Injury-related data and participant-related data (e.g., age, sex, diagnosed diseases, and medications) were collected from hospital records. The most common pre-injury diseases were circulatory (39.4%-43.2%), neurological (23.7%-25.2%), and psychiatric (25.8%-27.5%) disorders. Alcohol abuse was present in 18.4%-26.8%. The most common medications were for cardiovascular (33.1%-36.6%), central nervous system (21.4%-30.8%), and blood clotting and anemia indications (21.5%-22.6%). Of the screened patients, only 2.5% met all the enrollment criteria. Age, neurological conditions, and psychiatric problems were the most common reasons for exclusion. Most of the patients sustaining an mTBI have some pre-injury diseases or conditions that could affect clinical outcome. By excluding patients with pre-existing conditions, the patients with known risk factors for poor outcome remain poorly studied.
Collapse
Affiliation(s)
| | - Grant L Iverson
- 2 Department of Physical Medicine and Rehabilitation, Harvard Medical School; Spaulding Rehabilitation Hospital; MassGeneral Hospital for Children Sports Concussion Program; Red Sox Foundation and Massachusetts General Hospital Home Base Program , Boston, Massachusetts
| | - Anneli Kataja
- 3 Department of Radiology, Tampere University Hospital , Tampere, Finland
| | - Antti Brander
- 3 Department of Radiology, Tampere University Hospital , Tampere, Finland
| | - Juha Öhman
- 4 Department of Neurosciences and Rehabilitation, Tampere University Hospital , Tampere, Finland
| | - Teemu M Luoto
- 5 Department of Neurosurgery, Tampere University Hospital , Tampere, Finland
| |
Collapse
|
45
|
Walhovd KB, Bjørnebekk A, Haabrekke K, Siqveland T, Slinning K, Nygaard E, Fjell AM, Due-Tønnessen P, Bjørnerud A, Moe V. Child neuroanatomical, neurocognitive, and visual acuity outcomes with maternal opioid and polysubstance detoxification. Pediatr Neurol 2015; 52:326-32.e1-3. [PMID: 25595574 DOI: 10.1016/j.pediatrneurol.2014.11.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/14/2014] [Accepted: 11/18/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND Maternal opioid and polysubstance use during pregnancy is associated with an increased risk of child neurocognitive and visual problems and neuroanatomical differences. We hypothesized that, in contrast to findings from a previous study of children born to mothers not detoxified, children born to detoxified mothers would not show gross neuroanatomical and neurocognitive differences. METHODS Mothers with opioid and polysusbstance abuse problems and their infants (n = 11 + 12) were recruited from residential treatment institutions. Comparison mothers and infants (n = 12 + 12) were recruited from child health centers. The studies were approved by the Regional Committee of Medical Research Ethics. Children had magnetic resonance imaging scanning, neurocognitive, and visual acuity testing at 4.5 years. Neuroanatomical, cognitive, and visual acuity characteristics were compared across groups by analysis of variance and general linear models. RESULTS There were no significant differences across groups in neuroanatomical volumes, or cortical thickness, area, or volume. There were no differences in general neurocognitive functioning, but significantly lower left eye visual acuity, and a trend toward lower binocular visual acuity, in the drug-exposed relative to the comparison group. CONCLUSIONS The present study does not demonstrate gross differences relative to a comparison group in neuroanatomical and general neurocognitive characteristics of children born to mothers with opioid and polysubstance abuse who were detoxified during pregnancy. However, visual acuity was significantly lower in the drug-exposed group, requiring attention. There is a pressing need for additional and larger studies of long-term and specific child outcomes in this at-risk group.
Collapse
Affiliation(s)
- Kristine B Walhovd
- Research Group for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway; Department of Physical Medicine and Rehabilitation, Unit of Neuropsychology, Oslo University Hospital, Oslo, Norway.
| | - Astrid Bjørnebekk
- Research Group for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway; Department of Physical Medicine and Rehabilitation, Unit of Neuropsychology, Oslo University Hospital, Oslo, Norway
| | - Kristin Haabrekke
- The Center for Child and Adolescent Mental Health, Eastern and Southern Norway, Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway
| | | | - Kari Slinning
- The Center for Child and Adolescent Mental Health, Eastern and Southern Norway, Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway
| | - Egil Nygaard
- Research Group for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway
| | - Anders M Fjell
- Research Group for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway; Department of Physical Medicine and Rehabilitation, Unit of Neuropsychology, Oslo University Hospital, Oslo, Norway
| | - Paulina Due-Tønnessen
- Research Group for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway; Department of Radiology, Section of Neuroradiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Atle Bjørnerud
- Research Group for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway; Intervention Center, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Vibeke Moe
- The Center for Child and Adolescent Mental Health, Eastern and Southern Norway, Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway
| |
Collapse
|
46
|
Wollman SC, Alhassoon OM, Stern MJ, Hall MG, Rompogren J, Kimmel CL, Perez-Figueroa AM. White matter abnormalities in long-term heroin users: a preliminary neuroimaging meta-analysis. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2015; 41:133-8. [PMID: 25664621 DOI: 10.3109/00952990.2014.985829] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Diffusion tensor imaging has been used to explore white matter changes in heroin-dependent patients; however, results have been inconsistent. OBJECTIVES The current study meta-analytically examines the neuroimaging findings of all studies published before 2014 using the novel technique of Effect Size Signed Differential Mapping (ES-SDM). METHODS Two independent investigators searched three databases for whole-brain voxel-based fractional anisotropy morphometric studies involving heroin use without comorbid polysubstance abuse. Of 59 initial primary studies, four met stringent inclusion criteria. RESULTS RESULTS from this preliminary analysis indicate that heroin abusers may have significant reductions in fractional anisotropy in the bilateral frontal sub-gyral regions extending from the limbic structures to the prefrontal association cortices, implicating damage to the cingulum and superior longitudinal fasciculus. Exploratory moderator analyses indicate that the potential damage in the left cingulate gyrus may increase with longer use and decrease after long-term abstinence. CONCLUSION These preliminary findings suggest that heroin abuse is significantly associated with damage to white matter integrity. These results are considered preliminary and analyses should be revisited with more primary studies focusing on either long- or short-term abuse as well as abstinence.
Collapse
Affiliation(s)
- Scott C Wollman
- California School of Professional Psychology , San Diego and
| | | | | | | | | | | | | |
Collapse
|
47
|
Kantarci K. Fractional anisotropy of the fornix and hippocampal atrophy in Alzheimer's disease. Front Aging Neurosci 2014; 6:316. [PMID: 25431558 PMCID: PMC4230048 DOI: 10.3389/fnagi.2014.00316] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 10/27/2014] [Indexed: 11/13/2022] Open
Abstract
Decrease in the directionality of water diffusion measured with fractional anisotropy (FA) on diffusion tensor imaging has been linked to loss of myelin and axons in the white matter. Fornix FA is consistently decreased in patients with mild cognitive impairment (MCI) and Alzheimer’s disease (AD). Furthermore, decreased fornix FA is one of the earliest MRI abnormalities observed in cognitively normal individuals who are at an increased risk for AD, such as in pre-symptomatic carriers of familial AD mutations and in pre-clinical AD. Reductions of FA at these early stages, which predicted the decline in memory function. Fornix carries the efferent projections from the CA1 and CA3 pyramidal neurons of the hippocampus and subiculum, connecting these structures to the septal nuclei, anterior thalamic nucleus, mammillary bodies, and medial hypothalamus. Fornix also carries the afferent cholinergic and GABAergic projections from the medial septal nuclei and the adjacent diagonal band back to the medial temporal lobe, interconnecting the core limbic structures. Because fornix carries the axons projecting from the hippocampus, integrity of the fornix is in-part linked to the integrity of the hippocampus. In keeping with that, fornix FA is reduced in subjects with hippocampal atrophy, correlating with memory function. The literature on FA reductions in the fornix in the clinical spectrum of AD from pre-symptomatic carriers of familial AD mutations to pre-clinical AD, MCI, and dementia stages is reviewed.
Collapse
Affiliation(s)
- Kejal Kantarci
- Department of Radiology, Mayo Clinic , Rochester, MN , USA
| |
Collapse
|
48
|
Roos A, Kwiatkowski MA, Fouche JP, Narr KL, Thomas KGF, Stein DJ, Donald KA. White matter integrity and cognitive performance in children with prenatal methamphetamine exposure. Behav Brain Res 2014; 279:62-7. [PMID: 25446763 DOI: 10.1016/j.bbr.2014.11.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 10/29/2014] [Accepted: 11/04/2014] [Indexed: 11/29/2022]
Abstract
There is emerging evidence on the harmful effects of prenatal methamphetamine (MA) exposure on the structure and function of the developing brain. However, few studies have assessed white matter structural integrity in the presence of prenatal MA exposure, and results are inconsistent. This investigation thus used diffusion tensor imaging (DTI) to investigate white matter microstructure and cognitive performance in a group of prenatal MA exposed (or MA) children and controls of similar age. Seventeen MA children and 15 healthy controls (aged 6-7 years) underwent DTI and assessment of motor function and general cognitive ability. Whole brain analyses of white matter structure were performed using FSL's tract-based spatial statistics comparing fractional anisotropy (FA), mean diffusivity (MD), radial diffusivity (RD) and axial diffusivity (AD). Mean diffusion values were extracted from white matter regions shown to differ across groups to determine whether variations in FA predicted cognitive performance. Analyses were controlled for maternal nicotine use. MA children showed significantly lower FA as well as higher MD, RD and AD in tracts that traverse striatal, limbic and frontal regions. Abnormal FA levels in MA children were significantly associated with poorer motor coordination and general cognitive ability sub-items that relate to aspects of executive function. Our findings suggest that, consistent with previous studies in older children, there are disruptions of white matter microstructural integrity in striatal, limbic and frontal regions of young MA exposed children, with prominent cognitive implications. Future longitudinal studies may clarify how prenatal MA exposure affects white matter structural connectivity at different stages of brain maturation.
Collapse
Affiliation(s)
- Annerine Roos
- SU/UCT MRC Unit on Anxiety and Stress Disorders, Department of Psychiatry, PO Box 19063, Tygerberg 7505, South Africa.
| | | | | | - Katherine L Narr
- University of California, Los Angeles, Department of Neurology, Los Angeles, United States
| | | | - Dan J Stein
- University of Cape Town, Department of Psychiatry & Mental Health, South Africa
| | - Kirsty A Donald
- University of Cape Town, Division of Developmental Paediatrics, South Africa
| |
Collapse
|
49
|
Unraveling the secrets of white matter--bridging the gap between cellular, animal and human imaging studies. Neuroscience 2014; 276:2-13. [PMID: 25003711 PMCID: PMC4155933 DOI: 10.1016/j.neuroscience.2014.06.058] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 06/25/2014] [Indexed: 12/20/2022]
Abstract
The CNS white matter makes up about half of the human brain, and with advances in human imaging it is increasingly becoming clear that changes in the white matter play a major role in shaping human behavior and learning. However, the mechanisms underlying these white matter changes remain poorly understood. Within this special issue of Neuroscience on white matter, recent advances in our knowledge of the function of white matter, from the molecular level to human imaging, are reviewed. Collaboration between fields is essential to understand the function of the white matter, but due to differences in methods and field-specific 'language', communication is often hindered. In this review, we try to address this hindrance by introducing the methods and providing a basic background to myelin biology and human imaging as a prelude to the other reviews within this special issue.
Collapse
|
50
|
Irner TB, Teasdale TW, Nielsen T, Vedal S, Olofsson M. Cognitive, emotional and social development in adolescents born to substance using women. Scand J Psychol 2014; 55:319-25. [PMID: 24828950 DOI: 10.1111/sjop.12134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Accepted: 03/11/2014] [Indexed: 11/27/2022]
Abstract
The aim of this article is to investigate the long-term developmental consequences of being born to a substance-using mother, focusing on cognitive functions, attention, emotional and social development. The longitudinal sample comprised 48 adolescents aged 12-16 at the time of follow-up assessments, which included the Wechsler Intelligence Scale for Children-III, the Test of Everyday Attention for Children, The Tower of London test and the Strengths and Difficulties Questionnaire. The adolescents scored significantly lower than the norms on Wechsler's subtests and Full-Scale IQ, and on The Everyday Attention test. There were few differences on the Strengths and Difficulties Questionnaire. The girls reported significantly more hyperactivity than the British norms, and the teachers reported higher impact scores in boys, compared to the British norms. Thus, the results on cognitive consequences of maternal substance use appear to be very substantial while the emotional and social consequences do not. The results suggest serious negative effects of substance exposure in utero on attention and cognitive functioning in general.
Collapse
Affiliation(s)
- Tina Birk Irner
- Department of Psychology, Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | | | | |
Collapse
|