1
|
Fabiano N, Wong S, Gupta A, Tran J, Bhambra N, Min KK, Dragioti E, Barbui C, Fiedorowicz JG, Gosling CJ, Cortese S, Gandhi J, Saraf G, Shorr R, Vigod SN, Frey BN, Delorme R, Solmi M. Safety of psychotropic medications in pregnancy: an umbrella review. Mol Psychiatry 2024:10.1038/s41380-024-02697-0. [PMID: 39266712 DOI: 10.1038/s41380-024-02697-0] [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: 05/17/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 09/14/2024]
Abstract
Weighing risks and benefits of the use of psychotropic medications during pregnancy remains a challenge worldwide. We systematically assessed the strength of associations between psychotropic medication use in pregnant people with mental disorders and various adverse health outcomes in both pregnant people and foetuses. Systematic reviews with meta-analyses of observational studies investigating the association between exposure to psychotropic medication in pregnancy and any adverse health outcomes were included. Credibility was graded into convincing, highly suggestive, suggestive, weak or not significant. Quality of the meta-analyses and of individual studies were assessed with A Measurement Tool to Assess Systematic Reviews 2 (AMSTAR 2) the Newcastle-Ottawa Scale (NOS), respectively. We considered 21 meta-analyses encompassing 17,290,755 participants (AMSTAR 2 high = 1, low = 12, or critically low = 8). Evidence was suggestive for: (1) preterm birth in pregnant people with either any mental disorder (equivalent odds ratio 1.62 (95% confidence interval 1.24-2.12) or depression (1.65 [1.34-2.02]) receiving antidepressants during any trimester of pregnancy; (2) small for gestational age for pregnant people with depression receiving a SSRI during any trimester of pregnancy (1.50 [1.19-1.90]); and (3) major congenital malformation (1.24 [1.09-1.40]) or cardiac malformations (1.28 [1.11-1.47]) in babies for pregnant people with depression or anxiety receiving paroxetine during first trimester of pregnancy. Additional associations were supported by weak evidence, or were not statistically significant. This umbrella review found no convincing or highly suggestive level of evidence of adverse health outcomes associated with psychotropic medication use in pregnant people with mental disorders.
Collapse
Affiliation(s)
- Nicholas Fabiano
- SCIENCES Lab, Department of Psychiatry, University of Ottawa, Ottawa, ON, Canada
| | - Stanley Wong
- SCIENCES Lab, Department of Psychiatry, University of Ottawa, Ottawa, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Arnav Gupta
- Department of Medicine, University of Calgary, Calgary, AB, Canada
- College of Public Health, Kent State University, Kent, OH, US
| | - Jason Tran
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Nishaant Bhambra
- Department of Family Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Kevin K Min
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Elena Dragioti
- Research Laboratory Psychology of Patients, Families & Health Professionals, Department of Nursing, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Corrado Barbui
- WHO Collaborating Centre for Research and Training in Mental Health and Service Evaluation, Department of Neuroscience, Biomedicine and Movement Sciences, Section of Psychiatry, University of Verona, Verona, Italy
| | - Jess G Fiedorowicz
- Department of Psychiatry, University of Ottawa, Ottawa, ON, Canada
- Department of Mental Health, The Ottawa Hospital, Ottawa, ON, Canada
- Ottawa Hospital Research Institute (OHRI) Clinical Epidemiology Program, University of Ottawa, Ottawa, ON, Canada
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Corentin J Gosling
- DysCo Laboratory, Université Paris Nanterre, F9200, Nanterre, France
- Laboratory of Psychopathology and Health Process, Université Paris Cité, F92000, Paris, France
- Centre for Innovation in Mental Health, School of Psychology, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - Samuele Cortese
- Centre for Innovation in Mental Health, School of Psychology, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
- Clinical and Experimental Sciences (CNS and Psychiatry), Faculty of Medicine, University of Southampton, Southampton, UK
- Solent NHS Trust, Southampton, UK
- Hassenfeld Children's Hospital at NYU Langone, New York University Child Study Center, New York, NY, USA
- DiMePRe-J-Department of Precision and Regenerative Medicine-Jonic Area, University of Bari "Aldo Moro", Bari, Italy
| | - Jasmine Gandhi
- Department of Psychiatry, University of Ottawa, Ottawa, ON, Canada
- Department of Mental Health, The Ottawa Hospital, Ottawa, ON, Canada
- Ottawa Hospital Research Institute (OHRI) Clinical Epidemiology Program, University of Ottawa, Ottawa, ON, Canada
| | - Gayatri Saraf
- Department of Psychiatry, University of Ottawa, Ottawa, ON, Canada
- Ottawa Hospital Research Institute (OHRI) Clinical Epidemiology Program, University of Ottawa, Ottawa, ON, Canada
- The Royal's Institute of Mental Health Research, Ottawa, ON, Canada
| | - Risa Shorr
- Library Services, The Ottawa Hospital, Ottawa, ON, Canada
| | - Simone N Vigod
- Department of Psychiatry, Women's, College Hospital and University of Toronto, Toronto, ON, Canada
| | - Benicio N Frey
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
- Women's Health Concerns Clinic, St. Joseph's Healthcare Hamilton, Hamilton, ON, Canada
| | - Richard Delorme
- Child and Adolescent Psychiatry Department, Robert Debré Hospital, APHP, University of Paris Cité, Paris, France
| | - Marco Solmi
- SCIENCES Lab, Department of Psychiatry, University of Ottawa, Ottawa, ON, Canada.
- Department of Mental Health, The Ottawa Hospital, Ottawa, ON, Canada.
- Ottawa Hospital Research Institute (OHRI) Clinical Epidemiology Program, University of Ottawa, Ottawa, ON, Canada.
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
- Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany.
| |
Collapse
|
2
|
Mills-Huffnagle SL, Zawatsky CN, Bryant G, Ebert M, Augusto CM, Sipe A, Horvath N, Nyland JE. Differences in withdrawal symptoms, microglia activity, and cognitive functioning in rats exposed to continuous low-dose heroin in-utero. Neurotoxicol Teratol 2024; 105:107385. [PMID: 39182528 PMCID: PMC11403577 DOI: 10.1016/j.ntt.2024.107385] [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: 04/23/2024] [Revised: 07/26/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
INTRODUCTION Opioid use during pregnancy and subsequent neonatal opioid withdrawal syndrome (NOWS) have been associated with poor developmental outcomes including cognitive functioning. Less is known about the underlying molecular effects of prenatal opioid exposure and subsequent withdrawal; however, given the recent increase in NOWS cases, there is a pressing need to better understand these effects, which may partially explain cognitive deficits that have been observed in both preclinical NOWS models and patients with NOWS. This study evaluated the effects of prenatal heroin exposure and subsequent precipitated withdrawal symptoms on microglial reactivity in the nucleus accumbens (NAc), dorsal hippocampus (HC), and ventral tegmental area (VTA) in rat neonates, as well as cognitive functioning at three developmental time points using the Morris Water Maze (MWM) task. METHODS Heroin or saline (2 mg/kg) was randomly assigned and administered to six pregnant Sprague Dawley rat dams via osmotic minipump. A total of 63 rat neonates underwent naloxone-precipitated (5 mg/kg, subcutaneous injection) withdrawal testing at postnatal day 10 (PN10). Following withdrawal testing, neonates were randomly assigned to undergo perfusion and subsequent immunohistochemistry experiments to fluoresce Iba-1 for microglia detection, or to undergo the MWM task at three separate developmental time points (PN21-23; PN37; PN60) for cognitive testing. RESULTS Results suggest that in-utero heroin exposure led to an increase in ultrasonic vocalizations during naloxone-precipitated withdrawal; a sensitive index of withdrawal in rat neonates. Additional results suggest increased microglial reactivity in the HC and VTA, but not the NAc, as well as reduced performance during the MWM in the group exposed to heroin in-utero. DISCUSSION Together, these data suggest that in-utero opioid exposure is associated with microglial reactivity in brain regions associated with learning and memory, and may be associated with later cognitive deficits. Further research is needed to characterize these findings, which may inform future therapeutic strategies for this vulnerable population.
Collapse
Affiliation(s)
- Sara L Mills-Huffnagle
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences, 500 University Drive, Hershey, PA 17033, United States of America.
| | - Charles N Zawatsky
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences, 500 University Drive, Hershey, PA 17033, United States of America
| | - Gjhvona Bryant
- The Pennsylvania State University College of Medicine, Anatomy Graduate Program, 500 University Drive, Hershey, PA 17033, United States of America
| | - Michael Ebert
- The Pennsylvania State University College of Medicine, Anatomy Graduate Program, 500 University Drive, Hershey, PA 17033, United States of America
| | - Corinne M Augusto
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences, 500 University Drive, Hershey, PA 17033, United States of America
| | - Ann Sipe
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences, 500 University Drive, Hershey, PA 17033, United States of America
| | - Nelli Horvath
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences, 500 University Drive, Hershey, PA 17033, United States of America
| | - Jennifer E Nyland
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences, 500 University Drive, Hershey, PA 17033, United States of America
| |
Collapse
|
3
|
Nyberg H, Bogen IL, Nygaard E, Andersen JM. Effects of prenatal exposure to methadone or buprenorphine and maternal separation on anxiety-like behavior in rats. Drug Alcohol Depend 2024; 262:111367. [PMID: 39003831 DOI: 10.1016/j.drugalcdep.2024.111367] [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: 03/04/2024] [Revised: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND The use of medications for opioid use disorder such as methadone or buprenorphine is increasing among pregnant women. However, long-term effects of this treatment on the children's health are not well understood. A key challenge is distinguishing the effects of opioid exposure from other confounding factors associated with human opioid use, such as reduced maternal care. In this study, we therefore used a multi-risk factor design to examine anxiety-like behavior in rats prenatally exposed to methadone or buprenorphine, with or without maternal separation the first two weeks after birth. METHODS Female Sprague Dawley rats were exposed to methadone (10mg/kg/day), buprenorphine (1mg/kg/day) or sterile water throughout gestation. Half of the offspring in each litter experienced maternal separation for 3h per day from postnatal day 2 to 12. Male and female offspring (6-9 weeks) were tested in the open field, light-dark transition and elevated plus maze tests to assess anxiety-like behavior. RESULTS Offspring exposed to buprenorphine and not subjected to maternal separation displayed increased anxiety-like behavior in 3 out of 6 outcomes in the light-dark transition and elevated plus maze tests. Maternal separation did not exacerbate, but rather diminished this behavior. Males and females responded differently to methadone, with a trend towards reduced anxiety for males and increased anxiety for females. CONCLUSIONS Prenatal exposure to methadone or buprenorphine may increase the risk of developing anxiety-like behavior later in life, but the effect depends on specific subgroup characteristics. Further research is required to draw definitive conclusions.
Collapse
Affiliation(s)
- Henriette Nyberg
- Section of Forensic Research, Department of Forensic Sciences, Oslo University Hospital, P.O. Box 4950 Nydalen, Oslo 0424, Norway; Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, P.O. Box 1068 Blindern, Oslo 0316, Norway.
| | - Inger Lise Bogen
- Section of Forensic Research, Department of Forensic Sciences, Oslo University Hospital, P.O. Box 4950 Nydalen, Oslo 0424, Norway; Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, P.O. Box 1068 Blindern, Oslo 0316, Norway
| | - Egil Nygaard
- PROMENTA, Department of Psychology, Faculty of Social Sciences, University of Oslo, P.O. Box 1094 Blindern, Oslo 0317, Norway
| | - Jannike Mørch Andersen
- Section of Forensic Research, Department of Forensic Sciences, Oslo University Hospital, P.O. Box 4950 Nydalen, Oslo 0424, Norway; Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, P.O. Box 1068 Blindern, Oslo 0316, Norway
| |
Collapse
|
4
|
Tivnan P, Setty BN, Howard E, Agarwal J, Farris CW, Wachman EM, Castro-Aragon I. Ultrasound evaluation of brain parenchyma in preterm infants with prenatal opioid exposure. J Perinatol 2024; 44:1119-1124. [PMID: 37863985 DOI: 10.1038/s41372-023-01804-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 09/30/2023] [Accepted: 10/09/2023] [Indexed: 10/22/2023]
Abstract
OBJECTIVE To evaluate whether preterm infants with prenatal opioid exposure had differences in brain size on head ultrasounds (HUS) in comparison to non-exposed infants. STUDY DESIGN Preterm infants ≤34 weeks with prenatal opioid exposure (n = 47) and matched non-exposed infants (n = 62) with early HUSs were examined. Fifteen brain measurements were made and linear regression models performed to evaluate differences. RESULTS Brain measurements were smaller in the right ventricular index [β = -0.18 mm (95% CI -0.32, -0.03]), left ventricular index [β = -0.04 mm (95% CI -0.08, -0.003)], left basal ganglia insula [β = -0.10 mm (95% CI -0.15, -0.04)], right basal ganglia insula [β = -0.08 mm (95% CI -0.14, -0.03)], corpus callosum fastigium length [β = -0.16 mm (95% CI -0.25, -0.06)], intracranial height index [β = -0.31 mm (95% CI -0.44, -0.18)], and transcerebellar measurements [β = -0.13 (95% CI -0.25, -0.02)] in the opioid-exposed group. CONCLUSIONS Preterm infants with prenatal opioid exposure have smaller brain sizes compared to non-exposed infants, potentially increasing their risk for neurodevelopmental abnormalities.
Collapse
Affiliation(s)
- Patrick Tivnan
- Department of Radiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA.
- Department of Radiology, Johns Hopkins University, 4940 Eastern Avenue, Baltimore, MD, 21224, USA.
| | - Bindu N Setty
- Department of Pediatric Radiology, Boston University Chobanian & Avedisian School of Medicine, 715 Albany Street, Boston, MA, 02118, USA
| | - Eileen Howard
- Boston University Chobanian & Avedisian School of Medicine, 715 Albany Street, Boston, MA, 02118, USA
| | - Joel Agarwal
- Boston University School of Public Health, 715 Albany Street, Boston, MA, 02118, USA
| | - Chad W Farris
- Department of Pediatric Radiology, Boston University Chobanian & Avedisian School of Medicine, 715 Albany Street, Boston, MA, 02118, USA
| | - Elisha M Wachman
- Department of Pediatrics, Boston University Chobanian & Avedisian School of Medicine, 715 Albany Street, Boston, MA, 02118, USA
| | - Ilse Castro-Aragon
- Department of Pediatric Radiology, Boston University Chobanian & Avedisian School of Medicine, 715 Albany Street, Boston, MA, 02118, USA
| |
Collapse
|
5
|
Adegboyega O, Gayen Nee' Betal S, Urday P, Huang R, Bodycot K, Al-Kouatly HB, Solarin K, Chan JSY, Addya S, Boelig RC, Aghai ZH. DNA methylation patterns in umbilical cord blood from infants of methadone maintained opioid dependent mothers. Sci Rep 2024; 14:17298. [PMID: 39068260 PMCID: PMC11283475 DOI: 10.1038/s41598-024-66899-w] [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: 03/17/2024] [Accepted: 07/05/2024] [Indexed: 07/30/2024] Open
Abstract
Methadone maintenance treatment for opioid dependent mothers is standard of care. Infants of methadone maintained opioid dependent (MMOD) mothers have better outcomes compared to infants of opioid dependent mothers without treatment. However, when compared to non-exposed infants, infants of MMOD mothers are associated with worse outcomes. We conducted a pilot study to examine genome wide differential DNA methylation using cord blood samples from sixteen term and near-term infants of MMOD and opioid naïve mothers, excluding Infants with chorioamnionitis. A total of 152 differentially methylated loci were identified at a difference > + 2, < - 2 and p-value < 0.05. There were 90 hypermethylated loci (59 annotated genes) and 62 hypomethylated loci (38 annotated genes) observed. The hypermethylated and hypomethylated DNA changes involved multiple genes, pathways and networks that may explain some of the changes seen in infants of MMOD mothers. Top hypermethylated and hypomethylated genes involved areas of cell growth, neurodevelopment, vision and xenobiotic metabolism functions. Our data may explain the role of key pathways and genes relevant to neonatal outcomes seen from methadone exposure in pregnancy. Functional studies on the identified pathways and genes could lead to improved understanding of the mechanisms and identify areas for intervention.
Collapse
Affiliation(s)
- Oluwatobi Adegboyega
- Neonatology, Thomas Jefferson University, Attending Neonatologist, Nemours at TJU, Philadelphia, PA, USA
- Reilly Children's Hospital, Lehigh Valley Health Network, Allentown, USA
| | - Suhita Gayen Nee' Betal
- Neonatology, Thomas Jefferson University, Attending Neonatologist, Nemours at TJU, Philadelphia, PA, USA
| | - Pedro Urday
- Neonatology, Thomas Jefferson University, Attending Neonatologist, Nemours at TJU, Philadelphia, PA, USA
| | - Rachel Huang
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, USA
| | - Katherine Bodycot
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, USA
| | - Huda B Al-Kouatly
- Division of Maternal Fetal Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, USA
| | - Kolawole Solarin
- Neonatology, Thomas Jefferson University, Attending Neonatologist, Nemours at TJU, Philadelphia, PA, USA
| | - Joanna S Y Chan
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, USA
| | - Sankar Addya
- Laboratory of Cancer Genomics, Thomas Jefferson University, Philadelphia, USA
| | - Rupsa C Boelig
- Division of Maternal Fetal Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, USA
| | - Zubair H Aghai
- Neonatology, Thomas Jefferson University, Attending Neonatologist, Nemours at TJU, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Flores A, Nguyen NM, Devanaboyina M, Sanketh S, Athota P, Jagadesan S, Guda C, Yelamanchili SV, Pendyala G. Neurobehavioral Characterization of Perinatal Oxycodone-Exposed Offspring in Early Adolescence. J Neuroimmune Pharmacol 2024; 19:29. [PMID: 38874861 DOI: 10.1007/s11481-024-10129-7] [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: 01/05/2024] [Accepted: 05/25/2024] [Indexed: 06/15/2024]
Abstract
The opioid epidemic has received considerable attention, but the impact on perinatal opioid-exposed (POE) offspring remains underexplored. This study addresses the emerging public health challenge of understanding and treating POE children. We examined two scenarios using preclinical models: offspring exposed to oxycodone (OXY) in utero (IUO) and acute postnatal OXY (PNO). We hypothesized exposure to OXY during pregnancy primes offspring for neurodevelopmental deficits and severity of deficits is dependent on timing of exposure. Notable findings include reduced head size and brain weight in offspring. Molecular analyses revealed significantly lower levels of inflammasome-specific genes in the prefrontal cortex (PFC). Gene Set Enrichment Analysis (GSEA) and Ingenuity Pathway Analysis (IPA) highlighted the enrichment of genes associated with mitochondrial and synapse dysfunction in POE offspring. Western blot analysis validated IPA predictions of mitochondrial dysfunction in PFC-derived synaptosomes. Behavioral studies identified significant social deficits in POE offspring. This study presents the first comparative analysis of acute PNO- and IUO-offspring during early adolescence finding acute PNO-offspring have considerably greater deficits. The striking difference in deficit severity in acute PNO-offspring suggests that exposure to opioids in late pregnancy pose the greatest risk for offspring well-being.
Collapse
Affiliation(s)
- Adrian Flores
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, 68198, USA
- Department of Cellular and Integrative Physiology, UNMC, Omaha, NE, 68198, USA
| | - Nghi M Nguyen
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, 68198, USA
- Department of Genetics, Cell Biology and Anatomy, UNMC, Omaha, NE, 68198, USA
- Child Health Research Institute, Omaha, NE, 68198, USA
| | - Murali Devanaboyina
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, 68198, USA
| | - Samarth Sanketh
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, 68198, USA
| | - Pranavi Athota
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, 68198, USA
| | | | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, UNMC, Omaha, NE, 68198, USA
| | - Sowmya V Yelamanchili
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, 68198, USA
- Department of Genetics, Cell Biology and Anatomy, UNMC, Omaha, NE, 68198, USA
- National Strategic Research Institute, UNMC, Omaha, NE, USA
| | - Gurudutt Pendyala
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, 68198, USA.
- Department of Genetics, Cell Biology and Anatomy, UNMC, Omaha, NE, 68198, USA.
- Child Health Research Institute, Omaha, NE, 68198, USA.
- National Strategic Research Institute, UNMC, Omaha, NE, USA.
| |
Collapse
|
7
|
Tabachnick AR, Eiden RD, Labella MH, Dozier M. Effects of prenatal opioid exposure on infant sympathetic and parasympathetic nervous system activity. Psychophysiology 2024; 61:e14470. [PMID: 37888142 PMCID: PMC10939941 DOI: 10.1111/psyp.14470] [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: 03/10/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023]
Abstract
Prenatal opioid exposure has been associated with developmental problems, including autonomic nervous system dysregulation. However, little is known about the effects of prenatal opioid exposure on the autonomic nervous system beyond the first days of life, particularly across both the parasympathetic and sympathetic branches, and when accounting for exposure to other substances. The present study examined the effects of prenatal exposure to opioid agonist therapy (OAT, e.g., methadone) and other opioids on infant autonomic nervous system activity at rest and in response to a social stressor (the Still-Face Paradigm) at six months among 86 infants varying in prenatal opioid and other substance exposure. Results indicated that OAT and other opioids have unique effects on the developing autonomic nervous system that may further depend on subtype (i.e., methadone versus buprenorphine) and timing in gestation. Results are discussed in the context of theoretical models of the developing stress response system.
Collapse
Affiliation(s)
| | - Rina Das Eiden
- Department of Psychology, Pennsylvania State University, State College, Pennsylvania, USA
| | - Madelyn H Labella
- Department of Psychological Sciences, William & Mary, Williamsburg, Virginia, USA
| | - Mary Dozier
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware, USA
| |
Collapse
|
8
|
Abu YF, Singh S, Tao J, Chupikova I, Singh P, Meng J, Roy S. Opioid-induced dysbiosis of maternal gut microbiota during gestation alters offspring gut microbiota and pain sensitivity. Gut Microbes 2024; 16:2292224. [PMID: 38108125 PMCID: PMC10730209 DOI: 10.1080/19490976.2023.2292224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023] Open
Abstract
There has been a rapid increase in neonates born with a history of prenatal opioid exposure. How prenatal opioid exposure affects pain sensitivity in offspring is of interest, as this may perpetuate the opioid epidemic. While few studies have reported hypersensitivity to thermal pain, potential mechanisms have not been described. This study posits that alterations in the gut microbiome may underly hypersensitivity to pain in prenatally methadone-exposed 3-week-old male offspring, which were generated using a mouse model of prenatal methadone exposure. Fecal samples collected from dams and their offspring were subjected to 16s rRNA sequencing. Thermal and mechanical pain were assessed using the tail flick and Von Frey assays. Transcriptomic changes in whole brain samples of opioid or saline-exposed offspring were investigated using RNA-sequencing, and midbrain sections from these animals were subjected to qPCR profiling of genes related to neuropathic and inflammatory pain pathways. Prenatal methadone exposure increased sensitivity to thermal and mechanical pain and elevated serum levels of IL-17a. Taxonomical analysis revealed that prenatal methadone exposure resulted in significant alterations in fecal gut microbiota composition, including depletion of Lactobacillus, Bifidobacterium, and Lachnospiracea sp and increased relative abundance of Akkermansia, Clostridium sensu stricto 1, and Lachnoclostridium. Supplementation of the probiotic VSL#3 in dams rescued hypersensitivity to thermal and mechanical pain in prenatally methadone-exposed offspring. Similarly, cross-fostering prenatally methadone-exposed offspring to control dams also attenuated hypersensitivity to thermal pain in opioid-exposed offspring. Modulation of the maternal and neonatal gut microbiome with probiotics resulted in transcriptional changes in genes related to neuropathic and immune-related signaling in whole brain and midbrain samples of prenatally methadone-exposed offspring. Together, our work provides compelling evidence of the gut-brain-axis in mediating pain sensitivity in prenatally opioid-exposed offspring.
Collapse
Affiliation(s)
- Yaa F. Abu
- Department of Microbiology and Immunology, University of Miami, Miami, FL, USA
| | - Salma Singh
- Department of Surgery, University of Miami, Miami, FL, USA
| | - Junyi Tao
- Department of Surgery, University of Miami, Miami, FL, USA
| | | | - Praveen Singh
- Department of Surgery, University of Miami, Miami, FL, USA
| | - Jingjing Meng
- Department of Surgery, University of Miami, Miami, FL, USA
| | - Sabita Roy
- Department of Surgery, University of Miami, Miami, FL, USA
| |
Collapse
|
9
|
Hamilton R, Mulvihill A, Butler L, Chow A, Irving E, McCulloch DL, McNeil A, Michael K, Spowart KM, Waterson-Wilson J, Mactier H. Impaired vision in children prenatally exposed to methadone: an observational cohort study. Eye (Lond) 2024; 38:118-126. [PMID: 37402864 PMCID: PMC10764882 DOI: 10.1038/s41433-023-02644-3] [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: 04/03/2023] [Revised: 06/05/2023] [Accepted: 06/15/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND/OBJECTIVES To examine prevalence of failed visual assessment at 8-10 years in children born to methadone-maintained opioid dependent (MMOD) mothers and relate this to known in utero substance exposure. SUBJECTS/METHODS Follow up of observational cohort study of methadone-exposed and comparison children matched for birthweight, gestation and postcode of residence at birth. Participants were 144 children (98 exposed, 46 comparison). Prenatal drug exposure was previously established via comprehensive maternal and neonatal toxicology. Children were invited to attend for visual assessment and casenotes were reviewed. Presence of acuity poorer than 0.2 logMAR, strabismus, nystagmus and/or impaired stereovision constituted a 'fail'. Fail rates were compared between methadone-exposed and comparison children after adjusting for known confounding variables. RESULTS 33 children attended in person: data were also derived from casenote review for all children. After controlling for maternal reported tobacco use, methadone-exposed children were more likely to have a visual 'fail' outcome, adjusted odds ratio 2.6, 95% CI 1.1-6.2; adjusted relative risk 1.8 (95% CI 1.1-3.4). Visual 'fail' outcome rates did not differ between methadone-exposed children who had (n = 47) or had not (n = 51) received pharmacological treatment for neonatal abstinence/opioid withdrawal syndrome (NAS/NOWS); fail rate 62% vs 53% (95% CI of difference-11-27%). CONCLUSIONS Children born to MMOD mothers are almost twice as likely as unexposed peers to have significant visual abnormalities at primary school age. Prenatal methadone exposure should be considered in the differential diagnosis of nystagmus. Findings support visual assessment prior to school entry for children with any history of prenatal opioid exposure. TRIAL REGISTRATION The study was prospectively registered on ClinicalTrials.gov (NCT03603301), https://clinicaltrials.gov/ct2/show/NCT03603301 .
Collapse
Affiliation(s)
- R Hamilton
- Royal Hospital for Children, NHS Greater Glasgow & Clyde and the University of Glasgow, Glasgow, G51 4TF, UK.
| | - A Mulvihill
- Princess Alexandra Eye Pavilion, NHS Lothian, Edinburgh, EH3 9HA, UK
| | - L Butler
- Tennant Institute of Ophthalmology, NHS Greater Glasgow & Clyde, Glasgow, G12 0YN, UK
| | - A Chow
- School of Optometry and Vision Science, University of Waterloo, Waterloo, Ontario, N2L 3G1, Canada
| | - E Irving
- School of Optometry and Vision Science, University of Waterloo, Waterloo, Ontario, N2L 3G1, Canada
| | - D L McCulloch
- School of Optometry and Vision Science, University of Waterloo, Waterloo, Ontario, N2L 3G1, Canada
| | - A McNeil
- Royal Hospital for Children, NHS Greater Glasgow & Clyde, Glasgow, G51 4TF, UK
| | - K Michael
- Crosshouse Hospital, NHS Ayrshire & Arran, Kilmarnock, KA2 0BE, UK
| | - K M Spowart
- Specialist Children's Services, NHS Greater Glasgow & Clyde, Glasgow, G40 1DA, UK
| | - J Waterson-Wilson
- Royal Hospital for Children, NHS Greater Glasgow & Clyde, Glasgow, G51 4TF, UK
| | - H Mactier
- NHS Greater Glasgow & Clyde and the University of Glasgow, Glasgow, UK
| |
Collapse
|
10
|
Liu Y, Liu Y, Chen S, Kong Z, Guo Y, Wang H. Prenatal exposure to acetaminophen at different doses, courses and time causes testicular dysplasia in offspring mice and its mechanism. CHEMOSPHERE 2023; 345:140496. [PMID: 37865203 DOI: 10.1016/j.chemosphere.2023.140496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 10/08/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
Epidemiological investigation suggested that the use of acetaminophen during pregnancy may cause offspring testicular dysplasia, but no systematic study has been conducted. In this study, Kunming mice were given acetaminophen at different doses (100/200/400 mg/kg.d), courses (single/multiple), time (second/third trimester) during pregnancy. Fetal blood and testes were collected on gestaional day 18 for detection. The results indicated abnormal testicular development in the PAcE (prenatal acetaminophen exposure) groups. The maximum diameter/cross-sectional area decreased, the interstitial space widened, and decreased proliferation/increased apoptosis were observed, especially in the high-dose, multi-course and second-trimester groups. Meanwhile, the serum testosterone level decreased in PAcE groups, and the steroid synthesis function in Leydig cells, Sertoli and spermatogenic cell function were inhibited, it was more significant in high-dose, multi-course and second-trimester groups. Furthermore, Wnt signal pathway was activated but Notch signal pathway was inhibited in the PAcE groups. Finally, in vitro experiment, acetaminophen could inhibit spermatogonial cell proliferation, enhance apoptosis, and change Wnt/Notch signal pathway. In conclusion, this study confirmed that PAcE can change fetal testicular development in a dose, course and time-dependent manner, and found that multicellular function impaired. This study provides theoretical and experimental basis for systematically elucidating the developmental toxicity of acetaminophen in testis.
Collapse
Affiliation(s)
- Yi Liu
- Department of Pharmacology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Yi Liu
- Department of Pharmacology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China; Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Sijia Chen
- Department of Pharmacology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Ziyu Kong
- Department of Pharmacology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Yu Guo
- Department of Pharmacology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| | - Hui Wang
- Department of Pharmacology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
11
|
Yao H, Hu D, Wang J, Wu W, Zhao HH, Wang L, Gleeson J, Haddad GG. Buprenorphine and methadone differentially alter early brain development in human cortical organoids. Neuropharmacology 2023; 239:109683. [PMID: 37543137 DOI: 10.1016/j.neuropharm.2023.109683] [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: 04/10/2023] [Revised: 07/12/2023] [Accepted: 08/01/2023] [Indexed: 08/07/2023]
Abstract
Buprenorphine (BUP) and methadone (MTD) are used for medication-assisted treatment (MAT) in opioid use disorder. Although both medications show improved maternal and neonatal outcomes compared with illicit opioid use during pregnancy, BUP has exhibited more favorable outcomes to newborns than MTD. The underlying cellular and molecular mechanisms for the difference between BUP and MTD are largely unknown. Here, we examined the growth and neuronal activity in human cortical organoids (hCOs) exposed to BUP or MTD. We found that the growth of hCOs was significantly restricted in the MTD-treated but not in the BUP-treated hCOs and BUP attenuated the growth-restriction effect of MTD in hCOs. Furthermore, a κ-receptor agonist restricted while an antagonist alleviated the growth-restriction effect of MTD in hCOs. Since BUP is not only a μ-agonist but a κ-antagonist, the prevention of this growth-restriction by BUP is likely due to its κ-receptor-antagonism. In addition, using multielectrode array (MEA) technique, we discovered that both BUP and MTD inhibited neuronal activity in hCOs but BUP showed suppressive effects only at higher concentrations. Furthermore, κ-receptor antagonist nBNI did not prevent the MTD-induced suppression of neuronal activity in hCOs but the NMDA-antagonism of MTD (that BUP lacks) plays a role in the inhibition of neuronal activity. We conclude that, although both MTD and BUP are μ-opioid agonists, a) the additional κ-receptor antagonism of BUP mitigates the MTD-induced growth restriction during neurodevelopment and b) the lack of NMDA antagonism of BUP (in contrast to MTD) induces much less suppressive effect on neural network communications.
Collapse
Affiliation(s)
- Hang Yao
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Daisy Hu
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Juan Wang
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Wei Wu
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Helen H Zhao
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Lu Wang
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093, USA; Rady Children's Hospital, San Diego, CA, 92123, USA
| | - Joe Gleeson
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093, USA; Rady Children's Hospital, San Diego, CA, 92123, USA
| | - Gabriel G Haddad
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, 92093, USA; Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093, USA; Rady Children's Hospital, San Diego, CA, 92123, USA
| |
Collapse
|
12
|
Pande LJ, Arnet RE, Piper BJ. An Examination of the Complex Pharmacological Properties of the Non-Selective Opioid Modulator Buprenorphine. Pharmaceuticals (Basel) 2023; 16:1397. [PMID: 37895868 PMCID: PMC10610465 DOI: 10.3390/ph16101397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/24/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
The goal of this review is to provide a recent examination of the pharmacodynamics as well as pharmacokinetics, misuse potential, toxicology, and prenatal consequences of buprenorphine. Buprenorphine is currently a Schedule III opioid in the US used for opioid-use disorder (OUD) and as an analgesic. Buprenorphine has high affinity for the mu-opioid receptor (MOR), delta (DOR), and kappa (KOR) and intermediate affinity for the nociceptin (NOR). Buprenorphine's active metabolite, norbuprenorphine, crosses the blood-brain barrier, is a potent metabolite that attenuates the analgesic effects of buprenorphine due to binding to NOR, and is responsible for the respiratory depressant effects. The area under the concentration curves are very similar for buprenorphine and norbuprenorphine, which indicates that it is important to consider this metabolite. Crowding sourcing has identified a buprenorphine street value (USD 3.95/mg), indicating some non-medical use. There have also been eleven-thousand reports involving buprenorphine and minors (age < 19) at US poison control centers. Prenatal exposure to clinically relevant dosages in rats produces reductions in myelin and increases in depression-like behavior. In conclusion, the pharmacology of this OUD pharmacotherapy including the consequences of prenatal buprenorphine exposure in humans and experimental animals should continue to be carefully evaluated.
Collapse
Affiliation(s)
- Leana J. Pande
- Department of Medical Education, Geisinger Commonwealth School of Medicine, Scranton, PA 18509, USA; (L.J.P.); (R.E.A.)
- Touro College of Osteopathic Medicine, Middletown, NY 10027, USA
| | - Rhudjerry E. Arnet
- Department of Medical Education, Geisinger Commonwealth School of Medicine, Scranton, PA 18509, USA; (L.J.P.); (R.E.A.)
| | - Brian J. Piper
- Department of Medical Education, Geisinger Commonwealth School of Medicine, Scranton, PA 18509, USA; (L.J.P.); (R.E.A.)
- Center for Pharmacy Innovation and Outcomes, Danville, PA 17821, USA
| |
Collapse
|
13
|
Bann CM, Newman JE, Poindexter B, Okoniewski K, DeMauro S, Lorch SA, Wilson-Costello D, Ambalavanan N, Peralta-Carcelen M, Limperopoulos C, Kapse K, Davis JM, Walsh M, Merhar S. Outcomes of Babies with Opioid Exposure (OBOE): protocol of a prospective longitudinal cohort study. Pediatr Res 2023; 93:1772-1779. [PMID: 36042329 PMCID: PMC9971338 DOI: 10.1038/s41390-022-02279-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/01/2022] [Accepted: 08/05/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND While the health, social, and economic impacts of opioid addiction on adults and their communities are well known, the impact of maternal opioid use on the fetus exposed in utero is less well understood. METHODS This paper presents the protocol of the ACT NOW Outcomes of Babies with Opioid Exposure (OBOE) Study, a multi-site prospective longitudinal cohort study of infants with antenatal opioid exposure and unexposed controls. Study objectives are to determine the impact of antenatal opioid exposure on brain development and neurodevelopmental outcomes over the first 2 years of life and explore whether family, home, and community factors modify developmental trajectories during this critical time period. RESULTS Primary outcomes related to brain development include cortical volumes, deep cerebral gray matter volumes, resting-state functional connectivity measures, and structural connectivity measures using diffusion tensor imaging. Primary neurodevelopmental outcomes include visual abnormalities, cognitive, language, and motor skills measured using the Bayley Scales of Infant Development and social-emotional and behavioral problems and competence measured by the Brief Infant-Toddler Social and Emotional Assessment. CONCLUSIONS The OBOE study has been designed to overcome challenges of previous studies and will help further understanding of the effects of antenatal opioid exposure on early infant development. IMPACT This study will integrate MRI findings and comprehensive neurodevelopmental assessments to provide early insights into the functional topography of the brain in this high-risk population and assess MRI as a potential biomarker. Rather than conducting neuroimaging at a single time point, the study will include serial MRI assessments from birth to 2 years, allowing for the examination of trajectories throughout this period of rapid brain development. While previous studies often have had limited information on exposures, this study will use umbilical cord assays to accurately measure amounts of opioids and other substances from 20 weeks of gestation to birth.
Collapse
Affiliation(s)
- Carla M Bann
- RTI International, Research Triangle Park, NC, USA.
| | | | | | | | - Sara DeMauro
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Scott A Lorch
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | | | | | | | - Kushal Kapse
- Children's National Medical Center, Washington, DC, USA
| | | | - Michele Walsh
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Stephanie Merhar
- Cincinnati Children's Hospital Medical Center and University of Cincinnati Department of Pediatrics, Cincinnati, OH, USA
| |
Collapse
|
14
|
Bierce L, Tabachnick AR, Eiden RD, Dozier M, Labella MH. A 12-month follow-up of infant neurodevelopmental outcomes of prenatal opioid exposure and polysubstance use. Neurotoxicol Teratol 2023; 97:107176. [PMID: 37054901 PMCID: PMC10198960 DOI: 10.1016/j.ntt.2023.107176] [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: 12/01/2022] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 04/15/2023]
Abstract
BACKGROUND Prenatal opioid exposure has been associated with developmental deficits during infancy, but the literature is limited by simple group comparisons and lack of appropriate controls. Previously published research with the current sample documented unique associations between prenatal opioid exposure and developmental outcomes at three and six months, but less is known about associations later in infancy. METHOD The current study examined pre- and postnatal opioid and polysubstance exposure as predictors of parent-reported developmental status at 12 months of age. Participants were 85 mother-child dyads, oversampled for mothers taking opioid treatment medications during pregnancy. Maternal opioid and polysubstance use were reported using the Timeline Follow-Back Interview during the third trimester of pregnancy or up to one month postpartum and updated through the child's first year of life. Seventy-eight dyads participated in a 12-month assessment, including 68 with parent-reported developmental status on Ages and Stages Questionnaire. RESULTS At 12 months, average developmental scores fell within normal ranges and prenatal opioid exposure was not significantly related to any developmental outcomes. However, more prenatal alcohol exposure was significantly related to worse problem-solving scores, and this relationship remained after controlling for adjusted age and other substance exposure. CONCLUSION Although findings await replication with larger samples and more comprehensive measures, results suggest that unique developmental risks of prenatal opioid exposure may not persist through the first year of life. Effects of prenatal exposure to co-occurring teratogens, such as alcohol, may become apparent as children exposed to opioids develop.
Collapse
Affiliation(s)
- Lydia Bierce
- Department of Psychological Sciences, William and Mary, 540 Landrum Drive, Williamsburg, VA 23185, United States.
| | - Alexandra R Tabachnick
- Department of Medicine, University of Illinois at Chicago, 1200 W Harrison Street, Chicago, IL, 60607, United States
| | - Rina D Eiden
- Department of Psychology and the Social Science Research Institute, The Pennsylvania State University, 140 Moore Building, University Park, PA 16801, United States
| | - Mary Dozier
- Department of Psychological & Brain Sciences, University of Delaware, 108 Wolf Hall, Newark, DE 19717, United States
| | - Madelyn H Labella
- Department of Psychological Sciences, William and Mary, 540 Landrum Drive, Williamsburg, VA 23185, United States.
| |
Collapse
|
15
|
Wu CY, Chen HH, Tao PL, Yuan ZF. Comparisons of stress-related neuronal activation induced by restraint in adult male rat offspring with prenatal exposure to buprenorphine, methadone, or morphine. CHINESE J PHYSIOL 2023; 66:65-72. [PMID: 37082994 DOI: 10.4103/cjop.cjop-d-23-00015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023] Open
Abstract
Prenatal opioid exposure may impede the development of adaptive responses to environmental stimuli by altering the stress-sensitive brain circuitry located at the paraventricular nucleus of the hypothalamus (PVH) and locus coeruleus (LC). Corticotropin-releasing factor (CRF) released from neurons in the PVH has emerged as a key molecule to initiate and integrate the stress response. Methadone (Meth) and buprenorphine (Bu) are two major types of synthetic opioid agonists for first-line medication-assisted treatment of opioid (e.g., morphine, Mor) use disorder in pregnant women. No studies have compared the detrimental effects of prenatal exposure to Meth versus Bu on the stress response of their offspring upon reaching adulthood. In this study, we aimed to compare stress-related neuronal activation in the PVH and LC induced by restraint (RST) stress in adult male rat offspring with prenatal exposure to the vehicle (Veh), Bu, Meth, or Mor. CFos-immunoreactive cells were used as an indicator for neuronal activation. We found that RST induced less neuronal activation in the Meth or Mor exposure groups compared with that in the Bu or Veh groups; no significant difference was detected between the Bu and Veh exposure groups. RST-induced neuronal activation was completely prevented by central administration of a CRF receptor antagonist (α-helical CRF9-41, 10 μg/3 μL) in all exposure groups, suggesting the crucial role of CRF in this stress response. In offspring without RST, central administration of CRF (0.5 μg/3 μL)-induced neuronal activation in the PVH and LC. CRF-induced neuronal activation was lessened in the Meth or Mor exposure groups compared with that in the Bu or Veh groups; no significant difference was detected between the Bu and Veh exposure groups. Moreover, RST- or CRF-induced neuronal activation in the Meth exposure group was comparable with that in the Mor exposure group. Further immunohistochemical analysis revealed that the Meth and Mor exposure groups displayed less CRF neurons in the PVH of offspring with or without RST compared with the Bu or Veh groups. Thus, stress-induced neuronal activation in the PVH and LC was well preserved in adult male rat offspring with prenatal exposure to Bu, but it was substantially lessened in those with prenatal exposure to Meth or Mor. Lowered neuronal activation found in the Meth or Mor exposure groups may be, at least in part, due to the reduction in the density of CRF neurons in the PVH.
Collapse
Affiliation(s)
- Chia-Yen Wu
- Department of Physiology, Tzu Chi University, Hualien, Taiwan
| | - Hwei-Hsien Chen
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Pao-Luh Tao
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Zung Fan Yuan
- Department of Physiology, Tzu Chi University; Master Program in Biomedical Sciences, School of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
16
|
Prenatal Opioid Exposure and Neurodevelopmental Outcomes. J Neurosurg Anesthesiol 2023; 35:142-146. [PMID: 36745179 DOI: 10.1097/ana.0000000000000876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 12/12/2022]
Abstract
The opioid epidemic in the United States has resulted in a significant increase in opioid use disorder among pregnant women and a concomitant increase in the incidence of neonatal opioid withdrawal syndrome. The long-term consequences of prenatal opioid exposure on neurodevelopmental outcomes are not fully understood. Animal studies indicate increased neuronal apoptosis and decreased neuronal proliferation and myelination with opioid exposure in-utero. Meta-analyses of human studies suggest decreased cognition and psychomotor performance in infancy and deficits in cognition and language in preschool. However, current studies have primarily focused on heroin or methadone exposure and have been limited by small sample size, inadequate comparison groups, and the inability to account for additional risk factors and exposures such as polysubstance abuse, poor prenatal care, neonatal withdrawal and treatment with opioids, and unsupportive home environment. Future studies should aim to better understand the potential impact of these confounding factors on the neurodevelopmental trajectory of exposed infants. This review discusses the up-to-date literature, current gaps in knowledge, and considerations for future studies in the arena of prenatal opioid exposure and neurodevelopmental outcomes.
Collapse
|
17
|
Simmons SC, Grecco GG, Atwood BK, Nugent FS. Effects of prenatal opioid exposure on synaptic adaptations and behaviors across development. Neuropharmacology 2023; 222:109312. [PMID: 36334764 PMCID: PMC10314127 DOI: 10.1016/j.neuropharm.2022.109312] [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: 09/09/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
In this review, we focus on prenatal opioid exposure (POE) given the significant concern for the mental health outcomes of children with parents affected by opioid use disorder (OUD) in the view of the current opioid crisis. We highlight some of the less explored interactions between developmental age and sex on synaptic plasticity and associated behavioral outcomes in preclinical POE research. We begin with an overview of the rich literature on hippocampal related behaviors and plasticity across POE exposure paradigms. We then discuss recent work on reward circuit dysregulation following POE. Additional risk factors such as early life stress (ELS) could further influence synaptic and behavioral outcomes of POE. Therefore, we include an overview on the use of preclinical ELS models where ELS exposure during key critical developmental periods confers considerable vulnerability to addiction and stress psychopathology. Here, we hope to highlight the similarity between POE and ELS on development and maintenance of opioid-induced plasticity and altered opioid-related behaviors where similar enduring plasticity in reward circuits may occur. We conclude the review with some of the limitations that should be considered in future investigations. This article is part of the Special Issue on 'Opioid-induced addiction'.
Collapse
Affiliation(s)
- Sarah C Simmons
- Department of Pharmacology and Molecular Therapeutics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Greg G Grecco
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA; Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Brady K Atwood
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Fereshteh S Nugent
- Department of Pharmacology and Molecular Therapeutics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
18
|
Spowart KM, Reilly K, Mactier H, Hamilton R. Executive functioning, behavioural, emotional, and cognitive difficulties in school-aged children prenatally exposed to methadone. Front Pediatr 2023; 11:1118634. [PMID: 37144152 PMCID: PMC10151701 DOI: 10.3389/fped.2023.1118634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/22/2023] [Indexed: 05/06/2023] Open
Abstract
Aim The aim of this study was to examine executive function and emotional and behavioural difficulties of children aged between 8 and 10 years who had been prenatally exposed to methadone, compared to non-exposed peers. Methods Prospective study: third follow-up of an original cohort of 153 children born to methadone-maintained opioid-dependent mothers 2008-2010: previous investigations were at 1-3 days and at 6-7 months of age. Carers completed the Strength and Difficulties Questionnaire (SDQ) and the Behaviour Rating Inventory of Executive Function, Second Edition (BRIEF®2). Results were compared between exposed and non-exposed groups. Results Carers of 33 of 144 traceable children completed the measures. SDQ responses showed no group differences on subscales of emotional symptoms, conduct problems, or peer relationship problems. A marginally higher proportion of exposed children had a high or very high hyperactivity subscale score. Exposed children scored significantly higher on BRIEF®2 behavioural, emotional, and cognitive regulation indices, and on the global executive composite. After controlling for potentially confounding higher reported maternal tobacco use in the exposed group via regression modelling, the effect of methadone exposure reduced. Interpretation This study supports evidence that methadone exposure in utero is associated with adverse neurodevelopmental outcomes in childhood. Challenges in studying this population include difficulties with long-term follow-up and controlling for potentially confounding factors. Further investigation of the safety of methadone and other opioids in pregnancy must include consideration of maternal tobacco use.
Collapse
Affiliation(s)
- Katherine M. Spowart
- Specialist Children’s Services, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
| | - Kasey Reilly
- Specialist Children’s Services, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
| | - Helen Mactier
- Princess Royal Maternity, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Ruth Hamilton
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
- Department of Clinical Physics and Bioengineering, Royal Hospital for Children, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
- Correspondence: Ruth Hamilton
| |
Collapse
|
19
|
Minakova E, Mikati MO, Madasu MK, Conway SM, Baldwin JW, Swift RG, McCullough KB, Dougherty JD, Maloney SE, Al-Hasani R. Perinatal oxycodone exposure causes long-term sex-dependent changes in weight trajectory and sensory processing in adult mice. Psychopharmacology (Berl) 2022; 239:3859-3873. [PMID: 36269379 DOI: 10.1007/s00213-022-06257-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 10/03/2022] [Indexed: 11/28/2022]
Abstract
RATIONALE In utero opioid exposure is associated with lower weight and a neonatal opioid withdrawal syndrome (NOWS) at birth, along with longer-term adverse neurodevelopmental outcomes and mood disorders. While NOWS is sometimes treated with continued opioids, clinical studies have not addressed if long-term neurobehavioral outcomes are worsened with continued postnatal exposure to opioids. In addition, pre-clinical studies comparing in utero only opioid exposure to continued post-natal opioid administration for withdrawal mitigation are lacking. OBJECTIVES Here, we sought to understand the impact of continued postnatal opioid exposure on long term behavioral consequences. METHODS We implemented a rodent perinatal opioid exposure model of oxycodone (Oxy) exposure that included Oxy exposure until birth (short Oxy) and continued postnatal opioid exposure (long Oxy) spanning gestation through birth and lactation. RESULTS Short Oxy exposure was associated with a sex-specific increase in weight gain trajectory in adult male mice. Long Oxy exposure caused an increased weight gain trajectory in adult males and alterations in nociceptive processing in females. Importantly, there was no evidence of long-term social behavioral deficits, anxiety, hyperactivity, or memory deficits following short or long Oxy exposure. CONCLUSIONS Our findings suggest that offspring with prolonged opioid exposure experienced some long-term sequelae compared to pups with opioid cessation at birth. These results highlight the potential long-term consequences of opioid administration as a mitigation strategy for clinical NOWS symptomology and suggest alternatives should be explored.
Collapse
Affiliation(s)
- Elena Minakova
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Marwa O Mikati
- Department of Psychiatry, Washington University School of Medicine, Campus Box 8232, 660 South Euclid Avenue, St. Louis, MO, 63110-1093, USA.,Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA.,Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA.,Washington University Pain Management Center, Washington University School of Medicine, St. Louis, MO, USA.,Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA
| | - Manish K Madasu
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA.,Washington University Pain Management Center, Washington University School of Medicine, St. Louis, MO, USA.,Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA
| | - Sineadh M Conway
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA.,Washington University Pain Management Center, Washington University School of Medicine, St. Louis, MO, USA.,Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA
| | - Justin W Baldwin
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA.,Department of Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Raylynn G Swift
- Department of Psychiatry, Washington University School of Medicine, Campus Box 8232, 660 South Euclid Avenue, St. Louis, MO, 63110-1093, USA.,Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Katherine B McCullough
- Department of Psychiatry, Washington University School of Medicine, Campus Box 8232, 660 South Euclid Avenue, St. Louis, MO, 63110-1093, USA.,Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Joseph D Dougherty
- Department of Psychiatry, Washington University School of Medicine, Campus Box 8232, 660 South Euclid Avenue, St. Louis, MO, 63110-1093, USA.,Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA.,Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Susan E Maloney
- Department of Psychiatry, Washington University School of Medicine, Campus Box 8232, 660 South Euclid Avenue, St. Louis, MO, 63110-1093, USA. .,Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, USA.
| | - Ream Al-Hasani
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA. .,Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA. .,Washington University Pain Management Center, Washington University School of Medicine, St. Louis, MO, USA. .,Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
20
|
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: 3.0] [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
|
21
|
Gamble ME, Marfatia R, Diaz MR. Prenatal methadone exposure leads to long‐term memory impairments and disruptions of dentate granule cell function in a sex‐dependent manner. Addict Biol 2022; 27:e13215. [DOI: 10.1111/adb.13215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 12/16/2022]
Affiliation(s)
- Meredith E. Gamble
- Psychology Department Binghamton University 4000 Vestal Parkway E Binghamton NY 13902 USA
| | - Rhea Marfatia
- Psychology Department Binghamton University 4000 Vestal Parkway E Binghamton NY 13902 USA
| | - Marvin R. Diaz
- Psychology Department Binghamton University 4000 Vestal Parkway E Binghamton NY 13902 USA
| |
Collapse
|
22
|
Khoshnood-Shariaati M, Ashrafzadeh S, Dastjani-Farahani A, Zamani R, Naseh A. Comparison of Ocular and Brain Abnormalities Among Neonates With In Utero Exposure to Opium or Other Drugs. Cureus 2022; 14:e27648. [PMID: 36072164 PMCID: PMC9439622 DOI: 10.7759/cureus.27648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2022] [Indexed: 11/18/2022] Open
Abstract
Background There are growing concerns regarding ocular and brain abnormalities in infants who had in utero exposure to various kinds of substances/drugs. We compared the ocular and brain abnormalities among neonates based on the type of drug used by mothers. Methodology This prospective cohort study of 305 neonates included all neonates at Mahdieh Hospital, Tehran, Iran, who had their records for ophthalmic screening and brain sonography and were born to mothers with a history of substance use disorder (2014-2017). Demographic data, results for viral antibodies (human immunodeficiency virus, hepatitis C, and hepatitis B), and Apgar scores at one and five minutes were collected. We excluded neonates with Apgar score <8 at one minute, weight <1,800 g, gestational age <35 weeks, asphyxia, or anomalies. The neonates’ eyes were examined using tropicamide 0.5%, phenylephrine 2.5%, and tetracaine. Results The prevalence of substance use disorder among pregnant women was 1.8%. The study included 305 neonates with a mean gestational age of 37.8 ± 1.6 weeks, while the mean age of their mothers with substance use disorder was 29.8 ± 6.4 years. Ophthalmologic examination showed that 37 (12%) neonates had abnormal incomplete retina vascularization, and brain abnormalities were seen in 29 (9.5%) neonates; however, no difference was identified based on the type of drug used by mothers. The birth weight (BW) of the neonates depended on the type of drugs used by the mothers (p = 0.027). Maternal use of cannabis and amphetamine were associated with the lowest and highest BWs (2,800 ± 283 and 3,750 ± 42 g), respectively. Conclusions The BW of neonates depended on the type of drugs used by the mothers, where cannabis and amphetamine use were associated with the lowest and highest BWs, respectively. However, our data could not identify if neonates’ ocular and brain abnormalities differed based on the types of drugs. This study highlights the importance of a drug-free pregnancy and the need for addiction-prevention programs provided to women of childbearing age.
Collapse
|
23
|
Kinsella M, Capel Y, Nelson SM, Kearns RJ. Opioid substitution in pregnancy a narrative review: contemporary evidence for use of methadone and buprenorphine in pregnancy. JOURNAL OF SUBSTANCE USE 2022. [DOI: 10.1080/14659891.2022.2106600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
- M. Kinsella
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | - Y. Capel
- Foundation Programme, NHS Greater Glasgow and Clyde, Glasgow, UK
| | | | - R. J. Kearns
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| |
Collapse
|
24
|
Coluzzi F, Rullo L, Scerpa MS, Losapio LM, Rocco M, Billeci D, Candeletti S, Romualdi P. Current and Future Therapeutic Options in Pain Management: Multi-mechanistic Opioids Involving Both MOR and NOP Receptor Activation. CNS Drugs 2022; 36:617-632. [PMID: 35616826 PMCID: PMC9166888 DOI: 10.1007/s40263-022-00924-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/18/2022] [Indexed: 12/24/2022]
Abstract
Opioids are widely used in chronic pain management, despite major concerns about their risk of adverse events, particularly abuse, misuse, and respiratory depression from overdose. Multi-mechanistic opioids, such as tapentadol and buprenorphine, have been widely studied as a valid alternative to traditional opioids for their safer profile. Special interest was focused on the role of the nociceptin opioid peptide (NOP) receptor in terms of analgesia and improved tolerability. Nociceptin opioid peptide receptor agonists were shown to reinforce the antinociceptive effect of mu opioid receptor (MOR) agonists and modulate some of their adverse effects. Therefore, multi-mechanistic opioids involving both MOR and NOP receptor activation became a major field of pharmaceutical and clinical investigations. Buprenorphine was re-discovered in a new perspective, as an atypical analgesic and as a substitution therapy for opioid use disorders; and buprenorphine derivatives have been tested in animal models of nociceptive and neuropathic pain. Similarly, cebranopadol, a full MOR/NOP receptor agonist, has been clinically evaluated for its potent analgesic efficacy and better tolerability profile, compared with traditional opioids. This review overviews pharmacological mechanisms of the NOP receptor system, including its role in pain management and in the development of opioid tolerance. Clinical data on buprenorphine suggest its role as a safer alternative to traditional opioids, particularly in patients with non-cancer pain; while data on cebranopadol still require phase III study results to approve its introduction on the market. Other bifunctional MOR/NOP receptor ligands, such as BU08028, BU10038, and AT-121, are currently under pharmacological investigations and could represent promising analgesic agents for the future.
Collapse
Affiliation(s)
- Flaminia Coluzzi
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Polo Pontino, Latina, Italy
- Unit Anesthesia, Intensive Care and Pain Medicine, Sant'Andrea University Hospital, Rome, Italy
| | - Laura Rullo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Irnerio 48, Bologna, 40126, Italy
| | - Maria Sole Scerpa
- Unit Anesthesia, Intensive Care and Pain Medicine, Sant'Andrea University Hospital, Rome, Italy
| | - Loredana Maria Losapio
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Irnerio 48, Bologna, 40126, Italy
| | - Monica Rocco
- Department of Surgical and Medical Science and Translational Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Sanzio Candeletti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Irnerio 48, Bologna, 40126, Italy.
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Irnerio 48, Bologna, 40126, Italy
| |
Collapse
|
25
|
Jiang W, Merhar SL, Zeng Z, Zhu Z, Yin W, Zhou Z, Wang L, He L, Vannest J, Lin W. Neural alterations in opioid-exposed infants revealed by edge-centric brain functional networks. Brain Commun 2022; 4:fcac112. [PMID: 35602654 PMCID: PMC9117006 DOI: 10.1093/braincomms/fcac112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/29/2022] [Accepted: 05/03/2022] [Indexed: 12/02/2022] Open
Abstract
Prenatal opioid exposure has been linked to adverse effects spanning multiple neurodevelopmental domains, including cognition, motor development, attention, and vision. However, the neural basis of these abnormalities is largely unknown. A total of 49 infants, including 21 opioid-exposed and 28 controls, were enrolled and underwent MRI (43 ± 6 days old) after birth, including resting state functional MRI. Edge-centric functional networks based on dynamic functional connections were constructed, and machine-learning methods were employed to identify neural features distinguishing opioid-exposed infants from unexposed controls. An accuracy of 73.6% (sensitivity 76.25% and specificity 69.33%) was achieved using 10 times 10-fold cross-validation, which substantially outperformed those obtained using conventional static functional connections (accuracy 56.9%). More importantly, we identified that prenatal opioid exposure preferentially affects inter- rather than intra-network dynamic functional connections, particularly with the visual, subcortical, and default mode networks. Consistent results at the brain regional and connection levels were also observed, where the brain regions and connections associated with visual and higher order cognitive functions played pivotal roles in distinguishing opioid-exposed infants from controls. Our findings support the clinical phenotype of infants exposed to opioids in utero and may potentially explain the higher rates of visual and emotional problems observed in this population. Finally, our findings suggested that edge-centric networks could better capture the neural differences between opioid-exposed infants and controls by abstracting the intrinsic co-fluctuation along edges, which may provide a promising tool for future studies focusing on investigating the effects of prenatal opioid exposure on neurodevelopment.
Collapse
Affiliation(s)
- Weixiong Jiang
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Stephanie L. Merhar
- Perinatal Institute, Division of Neonatology, Cincinnati Children’s Hospital and University of Cincinnati Department of Pediatrics, Cincinnati OH, United States
| | - Zhuohao Zeng
- East Chapel Hill High School, Chapel Hill, North Carolina, United States
| | - Ziliang Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Weiyan Yin
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Zhen Zhou
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Li Wang
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Lili He
- Department of Radiology, Cincinnati Children’s Hospital and University of Cincinnati, Cincinnati OH, United States
| | - Jennifer Vannest
- Department of Communication Sciences and Disorders, University of Cincinnati, Cincinnati OH, United States
| | - Weili Lin
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| |
Collapse
|
26
|
Kinsella M, Halliday LOE, Shaw M, Capel Y, Nelson SM, Kearns RJ. Buprenorphine Compared with Methadone in Pregnancy: A Systematic Review and Meta-Analysis. Subst Use Misuse 2022; 57:1400-1416. [PMID: 35758300 DOI: 10.1080/10826084.2022.2083174] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
INTRODUCTION Illicit opioid use in pregnancy is associated with adverse maternal, neonatal, and childhood outcomes. Opioid substitution is recommended, but whether methadone or buprenorphine is the optimal agent remains unclear. METHODS We searched EMBASE, PubMed, Web of Science, Scopus, Open Gray, CINAHL and the Cochrane Central Registry of Controlled Trials (CENTRAL) from inception to April 2020 for randomized controlled trials (RCTs) and cohort studies comparing methadone and buprenorphine treatment for opioid-using mothers. Included studies assessed maternal and or neonatal outcomes. We used random-effects meta-analyses to estimate summary measures for outcomes and report these separately for RCTs and cohort studies. RESULTS Of 408 abstracts screened, 20 papers were included (4 RCTs, 16 cohort, 223 and 7028 participants respectively). All RCTs (4/4) had a high risk of bias and median (IQR) Newcastle Ottawa Scale for cohort studies was 7.5 (6-9). In both RCTs and cohort studies, buprenorphine was associated with; greater offspring birth weight (weighted mean difference [WMD] 343 g (95% CI: 40-645 g) in RCT and 184 g (95% CI: 121-247 g) in cohort studies); body length at birth (WMD 2.28 cm (95% CI: 1.06-3.49 cm) in RCTs and 0.65 cm (95% CI: 0.31-0.98 cm) in cohort studies); and reduced risk of prematurity (risk ratio [RR] 0.41 (95% CI: 0.18-0.93) in RCTs and 0.63 [95% CI: 0.53-0.75] in cohort studies) when compared to methadone. All other clinical outcomes were comparable. CONCLUSIONS Compared to methadone, buprenorphine was consistently associated with improved birthweight and gestational age, however given potential biases, results should be interpreted with caution.
Collapse
Affiliation(s)
- Michael Kinsella
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | - Lucy O E Halliday
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | | | | | - Scott M Nelson
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | - Rachel J Kearns
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| |
Collapse
|
27
|
Radhakrishnan R, Brown BP, Haas DM, Zang Y, Sparks C, Sadhasivam S. Pilot study of fetal brain development and morphometry in prenatal opioid exposure and smoking on fetal MRI. J Neuroradiol 2022; 49:53-58. [PMID: 33418054 PMCID: PMC8255323 DOI: 10.1016/j.neurad.2020.12.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 01/03/2023]
Abstract
PURPOSE The purpose of this study was to assess for any differences in brain maturation, structure and morphometry in fetuses exposed to opioids in utero, compared to non-opioid exposed fetuses on fetal MRI. METHODS We performed a prospective study in pregnant women using opioids and healthy pregnant women without prenatal opioid use. We evaluated brain maturation, structure, and morphometry on second or third trimester fetal MRI and assessed group differences. RESULTS 28 pregnant women were enrolled, 12 with opioid exposure (average gestational age 33.67, range 28-39 w), 9 of whom also smoked, and 16 without opioid exposure (average gestational age 32.53, range 27-38 w). There was a significant difference in the anteroposterior diameter of the fetal cerebellar vermis in the opioid exposed fetuses compared to non-opioid exposed fetuses (p = 0.004). There were no significant differences in brain biparietal diameter, fronto-occipital diameter, transverse cerebellar diameter and anteroposterior dimension of the pons in opioid exposed fetuses compared to non-opioid exposed fetuses. There were no abnormalities in brain maturation and no major brain structural abnormalities in the opioid exposed fetuses. CONCLUSION Smaller fetal anteroposterior cerebellar vermian dimension was associated with in utero opioid exposure. There were no abnormalities in brain maturation or major structural abnormalities in fetuses exposed to opioids.
Collapse
Affiliation(s)
- Rupa Radhakrishnan
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN
| | - Brandon P. Brown
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN
| | - David M. Haas
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN
| | - Yong Zang
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN
| | - Christina Sparks
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN
| | | |
Collapse
|
28
|
Merhar SL, Jiang W, Parikh NA, Yin W, Zhou Z, Tkach JA, Wang L, Kline-Fath BM, He L, Braimah A, Vannest J, Lin W. Effects of prenatal opioid exposure on functional networks in infancy. Dev Cogn Neurosci 2021; 51:100996. [PMID: 34388637 PMCID: PMC8363826 DOI: 10.1016/j.dcn.2021.100996] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 07/22/2021] [Accepted: 07/29/2021] [Indexed: 11/20/2022] Open
Abstract
Prenatal opioid exposure has been linked to altered neurodevelopment and visual problems such as strabismus and nystagmus. The neural substrate underlying these alterations is unclear. Resting-state functional connectivity MRI (rsfMRI) is an advanced and well-established technique to evaluate brain networks. Few studies have examined the effects of prenatal opioid exposure on resting-state network connectivity in infancy. In this pilot study, we characterized network connectivity in opioid-exposed infants (n = 19) and controls (n = 20) between 4–8 weeks of age using both a whole-brain connectomic approach and a seed-based approach. Prenatal opioid exposure was associated with differences in distribution of betweenness centrality and connection length, with positive connections unique to each group significantly longer than common connections. The unique connections in the opioid-exposed group were more often inter-network connections while unique connections in controls and connections common to both groups were more often intra-network. The opioid-exposed group had smaller network volumes particularly in the primary visual network, but similar network strength as controls. Network topologies as determined by dice similarity index were different between groups, particularly in visual and executive control networks. These results may provide insight into the neural basis for the developmental and visual problems associated with prenatal opioid exposure.
Collapse
Affiliation(s)
- Stephanie L Merhar
- Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital and University of Cincinnati, Department of Pediatrics, Cincinnati, OH, USA.
| | - Weixiong Jiang
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nehal A Parikh
- Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital and University of Cincinnati, Department of Pediatrics, Cincinnati, OH, USA
| | - Weiyan Yin
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zhen Zhou
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jean A Tkach
- Imaging Research Center, Cincinnati Children's Hospital, Cincinnati, OH, USA; Department of Radiology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Li Wang
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Beth M Kline-Fath
- Imaging Research Center, Cincinnati Children's Hospital, Cincinnati, OH, USA; Department of Radiology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Lili He
- Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital and University of Cincinnati, Department of Pediatrics, Cincinnati, OH, USA
| | - Adebayo Braimah
- Imaging Research Center, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Jennifer Vannest
- Department of Communication Sciences and Disorders, College of Allied Health Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Weili Lin
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
29
|
Labella MH, Eiden RD, Tabachnick AR, Sellers T, Dozier M. Infant neurodevelopmental outcomes of prenatal opioid exposure and polysubstance use. Neurotoxicol Teratol 2021; 86:107000. [PMID: 34116198 PMCID: PMC8277730 DOI: 10.1016/j.ntt.2021.107000] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 04/19/2021] [Accepted: 06/01/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Prenatal opioid exposure has been linked to adverse birth outcomes and delays in infant development. Existing literature is limited by a simple group-differences approach as well as inadequate controls for sociodemographic factors and polysubstance exposure co-occurring with prenatal opioid use. METHOD The current study assessed cumulative opioid exposure (duration of prescribed and illicit opioid use) as a predictor of infant birth outcomes and mother-reported developmental status at three and six months of age, controlling for polysubstance exposure. Participants were predominantly low-income pregnant and peripartum women, oversampled for mothers receiving medication-assisted treatment (MAT) for opioid use disorder. Prenatal opioid and non-opioid substance use were reported by mothers using a Timeline Follow-Back Interview completed during the third trimester and updated postnatally (infant age six months). RESULTS Developmental scores were in the normal range. However, total opioid exposure was positively related to premature birth and inversely related to mother-reported developmental status in specific domains. Associations with three-month fine motor skills and six-month communication skills were robust to controls for polysubstance exposure and sociodemographic covariates. CONCLUSIONS Results suggest unique effects of prenatal opioid exposure on the early development of fine motor and communication skills. Similar findings were obtained for prescribed and illicit opioid use, underscoring developmental risks of both MAT and untreated substance use. Exploratory analyses investigating type and timing of MAT suggest directions for future research.
Collapse
Affiliation(s)
- Madelyn H Labella
- Department of Psychological & Brain Sciences, University of Delaware, 108 Wolf Hall, Newark, DE 19716, United States.
| | - Rina D Eiden
- Department of Psychology, Penn State University, 140 Moore Building, University Park, PA 16801, United States
| | - Alexandra R Tabachnick
- Department of Psychological & Brain Sciences, University of Delaware, 108 Wolf Hall, Newark, DE 19716, United States
| | - Tabitha Sellers
- Department of Psychological & Brain Sciences, University of Delaware, 108 Wolf Hall, Newark, DE 19716, United States
| | - Mary Dozier
- Department of Psychological & Brain Sciences, University of Delaware, 108 Wolf Hall, Newark, DE 19716, United States
| |
Collapse
|
30
|
Carolien K, Annika M. Affective decision-making in children prenatally exposed to opioids. Scand J Psychol 2021; 62:529-536. [PMID: 34037260 DOI: 10.1111/sjop.12743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 04/01/2021] [Indexed: 11/29/2022]
Abstract
Although opioid maintenance therapy (OMT) is currently recommended for pregnant opioid-dependent women, potential effects on children's long-term development are still largely unknown. The current study assessed the long-term cognitive development of children born to women in OMT. Particularly, children's decision-making performance was assessed with a child-friendly version of the Iowa Gambling Task. Using a prospective longitudinal design, a cohort of children was followed from birth to middle childhood. Data were collected in Norway between 2005 and 2017. Participants included 41 children (aged 9-11 years), 20 of whom had histories of prenatal methadone or buprenorphine exposure. Background data were collected from personal interviews and medical records in 2005-2006. Children's affective decision-making was assessed in 2016-2017. Results showed no main effect of group on the net scores in the gambling task, F(1, 39) = 1.44, p = 0.24, η2 = 0.04, demonstrating no group differences in decision-making performance. A main effect of group was found on sensitivity to punishment, with children in the control group choosing the doors with the infrequent, but high punishment more often compared to children in the OMT group, F(1, 39) = 4.90, p = 0.03, η2 = 0.11. No main effect of group on decision-making speed was found, although results showed a significant interaction effect between group and gain, F(1, 8,194) = 4.09, p = 0.04, η2 = 0.001. Children prenatally exposed to opioids were found to have normal decision-making performance on an affective decision-making task and were able to consider future consequences when making decisions.
Collapse
Affiliation(s)
- Konijnenberg Carolien
- Department of Psychology, Inland Norway University of Applied Sciences, Lillehammer, Norway.,Cognitive Developmental Research Unit, Department of Psychology, University of Oslo, Oslo, Norway
| | - Melinder Annika
- Cognitive Developmental Research Unit, Department of Psychology, University of Oslo, Oslo, Norway.,Child- and Adolescents Mental Health, Oslo University hospital, Oslo, Norway
| |
Collapse
|
31
|
Wouldes TA, Crawford A, Stevens S, Stasiak K. Evidence for the Effectiveness and Acceptability of e-SBI or e-SBIRT in the Management of Alcohol and Illicit Substance Use in Pregnant and Post-partum Women. Front Psychiatry 2021; 12:634805. [PMID: 34025470 PMCID: PMC8131659 DOI: 10.3389/fpsyt.2021.634805] [Citation(s) in RCA: 7] [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: 11/28/2020] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
Alcohol and illicit psychoactive drug use during pregnancy have increased worldwide, putting women and their children's health and development at risk. Multiple drug use, comorbid psychiatric disorders, sexual and physical abuse are common in women who use alcohol and drugs during pregnancy. The effects on the mother include poor reproductive and life-long health, legal, family, and social problems. Additionally, the exposed child is at increased risk of long-term physical health, mental health, and developmental problems. The stigma associated with substance use during pregnancy and some clinicians' reticence to inquire about substance use means many women are not receiving adequate prenatal, substance abuse, and mental health care. Evidence for mHealth apps to provide health care for pregnant and post-partum women reveal the usability and effectiveness of these apps to reduce gestational weight gain, improve nutrition, promote smoking cessation and manage gestational diabetes mellitus, and treat depression and anxiety. Emerging evidence suggests mHealth technology using a public health approach of electronic screening, brief intervention, or referral to treatment (e-SBIRT) for substance use or abuse can overcome the typical barriers preventing women from receiving treatment for alcohol and drug use during pregnancy. This brief intervention delivered through a mobile device may be equally effective as SBIRT delivered by a health care professional in preventing maternal drug use, minimizing the effects to the exposed child, and providing a pathway to therapeutic options for a substance use disorder. However, larger studies in more diverse settings with women who have co-morbid mental illness and a constellation of social risk factors that are frequently associated with substance use disorders are needed.
Collapse
Affiliation(s)
- Trecia A. Wouldes
- Department of Psychological Medicine, Faculty of Medical and Health Science, University of Auckland, Auckland, New Zealand
| | - Andi Crawford
- Department of Psychological Medicine, Faculty of Medical and Health Science, University of Auckland, Auckland, New Zealand
- Te Ara Manapou, Parenting and Pregnancy Service, Hawke's Bay District Health Board, Hastings, New Zealand
| | - Suzanne Stevens
- Department of Psychological Medicine, Faculty of Medical and Health Science, University of Auckland, Auckland, New Zealand
| | - Karolina Stasiak
- Department of Psychological Medicine, Faculty of Medical and Health Science, University of Auckland, Auckland, New Zealand
| |
Collapse
|
32
|
Grecco GG, Mork BE, Huang JY, Metzger CE, Haggerty DL, Reeves KC, Gao Y, Hoffman H, Katner SN, Masters AR, Morris CW, Newell EA, Engleman EA, Baucum AJ, Kim J, Yamamoto BK, Allen MR, Wu YC, Lu HC, Sheets PL, Atwood BK. Prenatal methadone exposure disrupts behavioral development and alters motor neuron intrinsic properties and local circuitry. eLife 2021; 10:e66230. [PMID: 33724184 PMCID: PMC7993998 DOI: 10.7554/elife.66230] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/11/2021] [Indexed: 12/18/2022] Open
Abstract
Despite the rising prevalence of methadone treatment in pregnant women with opioid use disorder, the effects of methadone on neurobehavioral development remain unclear. We developed a translational mouse model of prenatal methadone exposure (PME) that resembles the typical pattern of opioid use by pregnant women who first use oxycodone then switch to methadone maintenance pharmacotherapy, and subsequently become pregnant while maintained on methadone. We investigated the effects of PME on physical development, sensorimotor behavior, and motor neuron properties using a multidisciplinary approach of physical, biochemical, and behavioral assessments along with brain slice electrophysiology and in vivo magnetic resonance imaging. Methadone accumulated in the placenta and fetal brain, but methadone levels in offspring dropped rapidly at birth which was associated with symptoms and behaviors consistent with neonatal opioid withdrawal. PME produced substantial impairments in offspring physical growth, activity in an open field, and sensorimotor milestone acquisition. Furthermore, these behavioral alterations were associated with reduced neuronal density in the motor cortex and a disruption in motor neuron intrinsic properties and local circuit connectivity. The present study adds to the limited body of work examining PME by providing a comprehensive, translationally relevant characterization of how PME disrupts offspring physical and neurobehavioral development.
Collapse
Affiliation(s)
- Gregory G Grecco
- Department of Pharmacology and Toxicology, Indiana University School of MedicineIndianapolisUnited States
- Indiana University School of Medicine, Medical Scientist Training ProgramIndianapolisUnited States
| | - Briana E Mork
- Department of Pharmacology and Toxicology, Indiana University School of MedicineIndianapolisUnited States
- Program in Medical Neuroscience, Stark Neurosciences Research Institute, Indiana University School of MedicineIndianapolisUnited States
| | - Jui-Yen Huang
- Department of Psychological and Brain Sciences, Indiana UniversityBloomingtonUnited States
- The Linda and Jack Gill Center for Biomolecular Sciences, Department of Psychological and Brain Science, Program in Neuroscience, Indiana UniversityBloomingtonUnited States
| | - Corinne E Metzger
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of MedicineIndianapolisUnited States
| | - David L Haggerty
- Department of Pharmacology and Toxicology, Indiana University School of MedicineIndianapolisUnited States
| | - Kaitlin C Reeves
- Department of Pharmacology and Toxicology, Indiana University School of MedicineIndianapolisUnited States
| | - Yong Gao
- Department of Pharmacology and Toxicology, Indiana University School of MedicineIndianapolisUnited States
| | - Hunter Hoffman
- Department of Pharmacology and Toxicology, Indiana University School of MedicineIndianapolisUnited States
| | - Simon N Katner
- Deparment of Psychiatry, Indiana University School of MedicineIndianapolisUnited States
| | - Andrea R Masters
- Clinical Pharmacology Analytical Core-Indiana University Simon Cancer Center, Indiana University School of MedicineIndianapolisUnited States
| | - Cameron W Morris
- Department of Pharmacology and Toxicology, Indiana University School of MedicineIndianapolisUnited States
- Department of Biology, Indiana University-Purdue UniversityIndianapolisUnited States
| | - Erin A Newell
- Deparment of Psychiatry, Indiana University School of MedicineIndianapolisUnited States
| | - Eric A Engleman
- Department of Pharmacology and Toxicology, Indiana University School of MedicineIndianapolisUnited States
| | - Anthony J Baucum
- Department of Pharmacology and Toxicology, Indiana University School of MedicineIndianapolisUnited States
- Department of Biology, Indiana University-Purdue UniversityIndianapolisUnited States
- Stark Neurosciences Research Institute, Indiana University School of MedicineIndianapolisUnited States
| | - Jiuen Kim
- Department of Pharmacology and Toxicology, Indiana University School of MedicineIndianapolisUnited States
- Stark Neurosciences Research Institute, Indiana University School of MedicineIndianapolisUnited States
| | - Bryan K Yamamoto
- Department of Pharmacology and Toxicology, Indiana University School of MedicineIndianapolisUnited States
- Stark Neurosciences Research Institute, Indiana University School of MedicineIndianapolisUnited States
| | - Matthew R Allen
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of MedicineIndianapolisUnited States
- Indiana Center for Musculoskeletal Health, Indiana University School of MedicineIndianapolisUnited States
| | - Yu-Chien Wu
- Stark Neurosciences Research Institute, Indiana University School of MedicineIndianapolisUnited States
- Department of Radiology and Imaging Sciences, Indiana University School of MedicineIndianapolisUnited States
| | - Hui-Chen Lu
- Department of Pharmacology and Toxicology, Indiana University School of MedicineIndianapolisUnited States
- Department of Psychological and Brain Sciences, Indiana UniversityBloomingtonUnited States
| | - Patrick L Sheets
- Department of Pharmacology and Toxicology, Indiana University School of MedicineIndianapolisUnited States
- Stark Neurosciences Research Institute, Indiana University School of MedicineIndianapolisUnited States
| | - Brady K Atwood
- Department of Pharmacology and Toxicology, Indiana University School of MedicineIndianapolisUnited States
- Stark Neurosciences Research Institute, Indiana University School of MedicineIndianapolisUnited States
| |
Collapse
|
33
|
Isaac L, van den Hoogen NJ, Habib S, Trang T. Maternal and iatrogenic neonatal opioid withdrawal syndrome: Differences and similarities in recognition, management, and consequences. J Neurosci Res 2021; 100:373-395. [PMID: 33675100 DOI: 10.1002/jnr.24811] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 02/01/2021] [Indexed: 11/12/2022]
Abstract
Opioids are potent analgesics used to manage pain in both young and old, but the increased use in the pregnant population has significant individual and societal implications. Infants dependent on opioids, either through maternal or iatrogenic exposure, undergo neonatal opioid withdrawal syndrome (NOWS), where they may experience withdrawal symptoms ranging from mild to severe. We present a detailed and original review of NOWS caused by maternal opioid exposure (mNOWS) and iatrogenic opioid intake (iNOWS). While these two entities have been assessed entirely separately, recognition and treatment of the clinical manifestations of NOWS overlap. Neonatal risk factors such as age, genetic predisposition, drug type, and clinical factors like type of opioid, cumulative dose of opioid exposure, and disease status affect the incidence of both mNOWS and iNOWS, as well as their severity. Recognition of withdrawal is dependent on clinical assessment of symptoms, and the use of clinical assessment tools designed to determine the need for pharmacotherapy. Treatment of NOWS relies on a combination of non-pharmacological therapies and pharmacological options. Long-term consequences of opioids and NOWS continue to generate controversy, with some evidence of anatomic brain changes, but conflicting animal and human clinical evidence of significant cognitive or behavioral impacts on school-age children. We highlight the current knowledge on clinically relevant recognition, treatment, and consequences of NOWS, and identify new advances in clinical management of the neonate. This review brings a unique clinical perspective and critically analyzes gaps between the clinical problem and our preclinical understanding of NOWS.
Collapse
Affiliation(s)
- Lisa Isaac
- Department of Anesthesia and Pain Medicine, Hospital for Sick Children, Toronto, ON, Canada.,Department of Anesthesiology and Pain Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Nynke J van den Hoogen
- Comparative Biology and Experimental Medicine, Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Toronto, ON, Canada
| | - Sharifa Habib
- Department of Neonatology, Hospital for Sick Children, Toronto, ON, Canada.,Lawrence S. Bloomberg Faculty of Nursing, University of Toronto, Toronto, ON, Canada
| | - Tuan Trang
- Comparative Biology and Experimental Medicine, Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Toronto, ON, Canada
| |
Collapse
|
34
|
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: 5.0] [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
|
35
|
Shannon J, Peters K, Blythe S. The Challenges to Promoting Attachment for Hospitalised Infants with NAS. CHILDREN-BASEL 2021; 8:children8020167. [PMID: 33671576 PMCID: PMC7926699 DOI: 10.3390/children8020167] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/18/2021] [Accepted: 02/18/2021] [Indexed: 11/16/2022]
Abstract
The postnatal period is crucial for infants in establishing a connection with and security in primary caregivers and can have enduring effects on attachment patterns. However, due to the need for symptom management, many infants diagnosed with neonatal abstinence syndrome (NAS) may be separated from primary caregivers and cared for in a neonatal intensive care unit (NICU) or special care nursery (SCN) soon after birth. Research has shown that substance-exposed infants are more likely to experience insecure attachment patterns with their primary caregivers and that mothers with a history of substance abuse are less sensitive to their infants’ cues. Therefore, the aim of this research was to explore nurses’ and midwives’ experiences in promoting the attachment relationship for infants admitted to an NICU/SCN with NAS. A qualitative research design was used to gather data on the experiences of nine nurses/midwives from various NICU and SCN settings in Australia. Individual, semi-structured interviews were conducted, and transcribed interviews were coded using thematic analysis. While nurses/midwives valued the attachment relationship for infants with NAS, facilitation of the attachment relationship was mainly promoted when the mother was present. However, parents were often reported to be absent from the nursery. Difficulties in promoting an attachment relationship were also identified when an infant had child protection involvement. This research identifies areas in need of innovative change regarding the approach taken to promote the attachment relationship for infants with NAS when they are admitted to an NICU/SCN.
Collapse
Affiliation(s)
- Jaylene Shannon
- Generalist Community Nursing, Mid North Coast Local Health District, Wauchope 2446, Australia;
| | - Kath Peters
- School of Nursing and Midwifery, Western Sydney University, Penrith 2750, Australia;
| | - Stacy Blythe
- School of Nursing and Midwifery, Western Sydney University, Penrith 2750, Australia;
- Correspondence:
| |
Collapse
|
36
|
Prenatal Opioid Exposure Enhances Responsiveness to Future Drug Reward and Alters Sensitivity to Pain: A Review of Preclinical Models and Contributing Mechanisms. eNeuro 2020; 7:ENEURO.0393-20.2020. [PMID: 33060181 PMCID: PMC7768284 DOI: 10.1523/eneuro.0393-20.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/01/2020] [Accepted: 10/05/2020] [Indexed: 12/21/2022] Open
Abstract
The opioid crisis has resulted in an unprecedented number of neonates born with prenatal opioid exposure (POE); however, the long-term effects of POE on offspring behavior and neurodevelopment remain relatively unknown. The advantages and disadvantages of the various preclinical POE models developed over the last several decades are discussed in the context of clinical and translational relevance. Although considerable and important variability exists among preclinical models of POE, the examination of these preclinical models has revealed that opioid exposure during the prenatal period contributes to maladaptive behavioral development as offspring mature including an altered responsiveness to rewarding drugs and increased pain response. The present review summarizes key findings demonstrating the impact of POE on offspring drug self-administration (SA), drug consumption, the reinforcing properties of drugs, drug tolerance, and other reward-related behaviors such as hypersensitivity to pain. Potential underlying molecular mechanisms which may contribute to this enhanced addictive phenotype in POE offspring are further discussed with special attention given to key brain regions associated with reward including the striatum, prefrontal cortex (PFC), ventral tegmental area (VTA), hippocampus, and amygdala. Improvements in preclinical models and further areas of study are also identified which may advance the translational value of findings and help address the growing problem of POE in clinical populations.
Collapse
|
37
|
Kongstorp M, Bogen IL, Steinsland S, Nerem E, Salih TW, Stiris T, Andersen JM. Prenatal exposure to methadone or buprenorphine alters µ-opioid receptor binding and downstream signaling in the rat brain. Int J Dev Neurosci 2020; 80:443-453. [PMID: 32484968 DOI: 10.1002/jdn.10043] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/14/2020] [Accepted: 05/27/2020] [Indexed: 11/11/2022] Open
Abstract
There is a growing concern related to the use of opioid maintenance treatment during pregnancy. Studies in both humans and animals have reported reduced cognitive functioning in offspring prenatally exposed to methadone or buprenorphine; however, little is known about the neurobiological mechanisms underlying these impairments. To reveal possible neurobiological effects of such in utero exposure, we examined brain tissue from methadone- and buprenorphine-exposed rat offspring previously shown to display impaired learning and memory. We studied µ-opioid receptor (MOR) and N-methyl-D-aspartate receptor (NMDAR) binding in the rat offspring cerebrum during development and in the hippocampus at young adulthood. Moreover, we examined activation of the Ca2+ /calmodulin-dependent protein kinase II (CaMKII) and the extracellular signal-regulated kinase (ERK), which are central in the downstream signaling of these receptors. The methadone- and buprenorphine-exposed rat pups displayed reduced MOR binding up to two weeks after birth, whereas the NMDAR binding was unaffected. Prenatal exposure to methadone or buprenorphine also resulted in decreased activation of CaMKII and/or ERK during development, while young adult offspring displayed increased hippocampal ERK activation. In conclusion, our findings suggest that prenatal exposure to exogenous opioids, such as methadone or buprenorphine, may disturb the endogenous opioid system during development, with long-term effects on proteins important for cognitive functioning.
Collapse
Affiliation(s)
- Mette Kongstorp
- Department of Forensic Sciences, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Inger Lise Bogen
- Department of Forensic Sciences, Oslo University Hospital, Oslo, Norway
- Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Synne Steinsland
- Department of Forensic Sciences, Oslo University Hospital, Oslo, Norway
| | - Elisabeth Nerem
- Department of Forensic Sciences, Oslo University Hospital, Oslo, Norway
| | | | - Tom Stiris
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Neonatal Intensive Care, Oslo University Hospital, Oslo, Norway
| | - Jannike Mørch Andersen
- Department of Forensic Sciences, Oslo University Hospital, Oslo, Norway
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|