1
|
Spodnick MB, McElderry SC, Diaz MR. Opioid receptor signaling throughout ontogeny: Shaping neural and behavioral trajectories. Neurosci Biobehav Rev 2025; 170:106033. [PMID: 39894419 PMCID: PMC11851333 DOI: 10.1016/j.neubiorev.2025.106033] [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: 12/05/2024] [Revised: 01/17/2025] [Accepted: 01/26/2025] [Indexed: 02/04/2025]
Abstract
Due to the recent and ongoing opioid crisis in the United States, exposure to opioid drugs in utero is becoming more common, including during medication-assisted therapy used to treat opioid use disorder. As such, careful consideration of opioidergic signaling in utero and beyond, as well as alterations to this signaling via introduction of exogenous opioids, is warranted. This review explores the ontogeny and function of the Mu, Kappa and Delta opioid receptor systems throughout the lifespan, highlighting their importance in guiding neurobehavioral development. We argue for a paradigm shift in conceptualization of opioids as not only contributors within their own system, but also vital regulators of a multitude of downstream neurodevelopmental processes.
Collapse
Affiliation(s)
- Mary B Spodnick
- Binghamton University, 4400 Vestal Parkway East, Binghamton, NY, USA.
| | | | - Marvin R Diaz
- Binghamton University, 4400 Vestal Parkway East, Binghamton, NY, USA.
| |
Collapse
|
2
|
McLaurin KA, Ott RK, Mactutus CF, Booze RM. Adolescent oral oxycodone self-administration disrupts neurobehavioral and neurocognitive development. Neuropharmacology 2024; 258:110064. [PMID: 38981578 PMCID: PMC11418068 DOI: 10.1016/j.neuropharm.2024.110064] [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: 02/22/2024] [Revised: 07/05/2024] [Accepted: 07/05/2024] [Indexed: 07/11/2024]
Abstract
Nonmedical use of prescription opioids peaks during late adolescence, a developmental period associated with the maturation of higher-order cognitive processes. To date, however, how chronic adolescent oxycodone (OXY) self-administration alters neurobehavioral (i.e., locomotion, startle reactivity) and/or neurocognitive (i.e., preattentive processes, intrasession habituation, stimulus-reinforcement learning, sustained attention) function has not yet been systematically evaluated. Hence, the rationale was built for establishing the dose-dependency of adolescent OXY self-administration on the trajectory of neurobehavioral and neurocognitive development. From postnatal day (PD) 35 to PD 105, an age in rats that corresponds to the adolescent and young adult period in humans, male and female F344/N rats received access to either oral OXY (0, 2, 5, or 10 mg/kg) or water under a two-bottle choice experimental paradigm. Independent of biological sex or dose, rodents voluntarily escalated their OXY intake across ten weeks. A longitudinal experimental design revealed prominent OXY-induced impairments in neurobehavioral development, characterized by dose-dependent increases in locomotion and sex-dependent increases in startle reactivity. Systematic manipulation of the interstimulus interval in prepulse inhibition supports an OXY-induced impairment in preattentive processes. Despite the long-term cessation of OXY intake, rodents with a history of chronic adolescent oral OXY self-administration exhibited deficits in sustained attention; albeit no alterations in stimulus-reinforcement learning were observed. Taken together, adolescent oral OXY self-administration induces selective long-term alterations in neurobehavioral and neurocognitive development enjoining the implementation of safer prescribing guidelines for this population.
Collapse
Affiliation(s)
- Kristen A McLaurin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40508, USA.
| | - Rachael K Ott
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, University of South Carolina, 1512 Pendleton Street, Columbia, SC, 29208, USA
| | - Charles F Mactutus
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, University of South Carolina, 1512 Pendleton Street, Columbia, SC, 29208, USA
| | - Rosemarie M Booze
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, University of South Carolina, 1512 Pendleton Street, Columbia, SC, 29208, USA
| |
Collapse
|
3
|
Myers AM, Wallin CM, Richardson LM, Duran J, Neole SR, Kulaglic N, Davidson C, Perrine SA, Bowen SE, Brummelte S. The effects of buprenorphine and morphine during pregnancy: Impact of exposure length on maternal brain, behavior, and offspring neurodevelopment. Neuropharmacology 2024; 257:110060. [PMID: 38960134 PMCID: PMC11285462 DOI: 10.1016/j.neuropharm.2024.110060] [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: 04/26/2024] [Revised: 06/21/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024]
Abstract
The escalating incidence of opioid-related issues among pregnant women in the United States underscores the critical necessity to understand the effects of opioid use and Medication for Opioid Use Disorders (MOUDs) during pregnancy. This research employed a translational rodent model to examine the impact of gestational exposure to buprenorphine (BUP) or morphine on maternal behaviors and offspring well-being. Female rats received BUP or morphine before conception, representing established use, with exposure continuing until postnatal day 2 or discontinued on gestational day 19 to mimic treatment cessation before birth. Maternal behaviors - including care, pup retrieval, and preference - as well as hunting behaviors and brain neurotransmitter levels were assessed. Offspring were evaluated for mortality, weight, length, milk bands, surface righting latency, withdrawal symptoms, and brain neurotransmitter levels. Our results reveal that regardless of exposure length (i.e., continued or discontinued), BUP resulted in reduced maternal care in contrast to morphine-exposed and control dams. Opioid exposure altered brain monoamine levels in the dams and offspring, and was associated with increased neonatal mortality, reduced offspring weight, and elevated withdrawal symptoms compared to controls. These findings underscore BUP's potential disruption of maternal care, contributing to increased pup mortality and altered neurodevelopmental outcomes in the offspring. This study calls for more comprehensive research into prenatal BUP exposure effects on the maternal brain and infant development with the aim to mitigate adverse outcomes in humans exposed to opioids during pregnancy.
Collapse
Affiliation(s)
- Abigail M Myers
- Department of Psychology, Wayne State University, Detroit, MI, 48202, USA
| | - Chela M Wallin
- Department of Psychology, Wayne State University, Detroit, MI, 48202, USA
| | | | - Jecenia Duran
- Department of Psychology, Wayne State University, Detroit, MI, 48202, USA
| | - Surbhi R Neole
- Department of Psychology, Wayne State University, Detroit, MI, 48202, USA
| | - Nejra Kulaglic
- Department of Psychology, Wayne State University, Detroit, MI, 48202, USA
| | - Cameron Davidson
- Dept of Behavioral Neuroscience and Psychiatry, Wayne State University, Detroit, MI, 48202, USA
| | - Shane A Perrine
- Dept of Behavioral Neuroscience and Psychiatry, Wayne State University, Detroit, MI, 48202, USA; Translational Neuroscience Program, Wayne State University, Detroit, MI, 48202, USA
| | - Scott E Bowen
- Department of Psychology, Wayne State University, Detroit, MI, 48202, USA
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, Detroit, MI, 48202, USA; Translational Neuroscience Program, Wayne State University, Detroit, MI, 48202, USA.
| |
Collapse
|
4
|
Asad M, Bonner-Reid FT, Aldoohan F, Marrelli LM, Ghanie N, Attia Hussein Mahmoud H, Venkatraj Srividya S, Devadas Gandhi P, Zehra M, Nazir Z. Reviewing the Impact of Maternal Opioid Use Disorder on Fetal Development and Long-Term Pediatric Health Outcomes. Cureus 2024; 16:e72192. [PMID: 39583351 PMCID: PMC11583522 DOI: 10.7759/cureus.72192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2024] [Indexed: 11/26/2024] Open
Abstract
Opioid use during pregnancy has emerged as a notable public health concern with far-reaching impacts on both maternal and child health. This review investigates the consequences of opioid use disorder (OUD) and maternal opioid use during pregnancy, focusing on fetal and pediatric outcomes and available treatment options. The study focuses on the rising prevalence of opioid use among pregnant women, with emphasis on the adverse effects on neonates, including neonatal abstinence syndrome (NAS), preterm birth, low birth weight, and long-term cognitive and behavioral deficits. Furthermore, it explores the neurological implications of opioid exposure on fetal brain development, emphasizing disrupted synaptic plasticity, oligodendrocyte differentiation, and myelination. Current treatment strategies such as opioid maintenance therapy (OMT) with methadone and buprenorphine are discussed, including their benefits in improving prenatal care adherence but also their associated risks, such as NAS and small birth weights. Maternal OUD underscores the need for a multidisciplinary approach to manage opioid dependence in pregnant women effectively and calls for more comprehensive research to address the long-term developmental impacts on children, as well as to explore more effective prevention and intervention strategies for maternal OUD.
Collapse
Affiliation(s)
- Manahil Asad
- Medicine and Surgery, Fauji Foundation Hospital-Foundation University Medical College (FUMC) Islamabad, Islamabad, PAK
| | - Felicia T Bonner-Reid
- Medicine and Surgery, University of Medical Sciences of Manzanillo (Celia Sánchez Manduley), Manzanillo, CUB
| | - Fawaz Aldoohan
- Family Medicine, American Academy of Research and Academics, Newark, USA
| | | | - Neisha Ghanie
- College of Medicine, American University of Antigua, Coolidge, ATG
| | | | | | - Preanka Devadas Gandhi
- Graduate Medical Education, Metropolitan University College of Medicine, St. John's, ATG
| | - Muneeza Zehra
- Internal Medicine, Karachi Medical and Dental College, Karachi, PAK
| | - Zahra Nazir
- Internal Medicine, Combined Military Hospital (CMH) Quetta, Quetta, PAK
| |
Collapse
|
5
|
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
|
6
|
Dwivedi I, Haddad GG. Investigating the neurobiology of maternal opioid use disorder and prenatal opioid exposure using brain organoid technology. Front Cell Neurosci 2024; 18:1403326. [PMID: 38812788 PMCID: PMC11133580 DOI: 10.3389/fncel.2024.1403326] [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: 03/19/2024] [Accepted: 05/01/2024] [Indexed: 05/31/2024] Open
Abstract
Over the past two decades, Opioid Use Disorder (OUD) among pregnant women has become a major global public health concern. OUD has been characterized as a problematic pattern of opioid use despite adverse physical, psychological, behavioral, and or social consequences. Due to the relapsing-remitting nature of this disorder, pregnant mothers are chronically exposed to exogenous opioids, resulting in adverse neurological and neuropsychiatric outcomes. Collateral fetal exposure to opioids also precipitates severe neurodevelopmental and neurocognitive sequelae. At present, much of what is known regarding the neurobiological consequences of OUD and prenatal opioid exposure (POE) has been derived from preclinical studies in animal models and postnatal or postmortem investigations in humans. However, species-specific differences in brain development, variations in subject age/health/background, and disparities in sample collection or storage have complicated the interpretation of findings produced by these explorations. The ethical or logistical inaccessibility of human fetal brain tissue has also limited direct examinations of prenatal drug effects. To circumvent these confounding factors, recent groups have begun employing induced pluripotent stem cell (iPSC)-derived brain organoid technology, which provides access to key aspects of cellular and molecular brain development, structure, and function in vitro. In this review, we endeavor to encapsulate the advancements in brain organoid culture that have enabled scientists to model and dissect the neural underpinnings and effects of OUD and POE. We hope not only to emphasize the utility of brain organoids for investigating these conditions, but also to highlight opportunities for further technical and conceptual progress. Although the application of brain organoids to this critical field of research is still in its nascent stages, understanding the neurobiology of OUD and POE via this modality will provide critical insights for improving maternal and fetal outcomes.
Collapse
Affiliation(s)
- Ila Dwivedi
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Gabriel G. Haddad
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, United States
- Department of Neurosciences, School of Medicine, University of California, San Diego, La Jolla, CA, United States
- Rady Children’s Hospital, San Diego, CA, United States
| |
Collapse
|
7
|
Myers AM, Bowen SE, Brummelte S. Maternal care behavior and physiology moderate offspring outcomes following gestational exposure to opioids. Dev Psychobiol 2023; 65:e22433. [PMID: 38010303 DOI: 10.1002/dev.22433] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/31/2023] [Accepted: 09/29/2023] [Indexed: 11/29/2023]
Abstract
The opioid epidemic has resulted in a drastic increase in gestational exposure to opioids. Opioid-dependent pregnant women are typically prescribed medications for opioid use disorders ("MOUD"; e.g., buprenorphine [BUP]) to mitigate the harmful effects of abused opioids. However, the consequences of exposure to synthetic opioids, particularly BUP, during gestation on fetal neurodevelopment and long-term outcomes are poorly understood. Further, despite the known adverse effects of opioids on maternal care, many preclinical and clinical studies investigating the effects of gestational opioid exposure on offspring outcomes fail to report on maternal care behaviors. Considering that offspring outcomes are heavily dependent upon the quality of maternal care, it is important to evaluate the effects of gestational opioid exposure in the context of the mother-infant dyad. This review compares offspring outcomes after prenatal opioid exposure and after reduced maternal care and integrates this information to potentially identify common underlying mechanisms. We explore whether adverse outcomes after gestational BUP exposure are due to direct effects of opioids in utero, deficits in maternal care, or a combination of both factors. Finally, suggestions for improving preclinical models of prenatal opioid exposure are provided to promote more translational studies that can help to improve clinical outcomes for opioid-dependent mothers.
Collapse
Affiliation(s)
- Abigail M Myers
- Department of Psychology, Wayne State University, Detroit, Michigan, USA
| | - Scott E Bowen
- Department of Psychology, Wayne State University, Detroit, Michigan, USA
- Translational Neuroscience Program, Wayne State University, Detroit, Michigan, USA
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, Detroit, Michigan, USA
- Translational Neuroscience Program, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
8
|
Flores A, Nguyen NM, Pendyala G. Developmental outcomes with perinatal exposure (DOPE) to prescription opioids. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2023; 2:339-351. [PMID: 38058996 PMCID: PMC10696573 DOI: 10.1515/nipt-2023-0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/07/2023] [Indexed: 12/08/2023]
Abstract
Researchers have found considerable evidence in the past 20 years that perinatal opioid exposure leads to an increased risk of developmental disorders in offspring that persist into adulthood. The use of opioids to treat pain concerning pregnancy, delivery, and postpartum complications has been rising. As a result, communities have reported a 300-400 % increase in Neonatal Opioid Withdrawal Syndrome (NOWS). NOWS represents the initial stage of several behavioral, phenotypic, and synaptic deficits. This review article summarizes the Developmental Outcomes of Perinatal Exposure (DOPE) to prescription opioids. Moreover, we also seek to connect these findings to clinical research that describes DOPE at multiple stages of life. Since specific mechanisms that underlie DOPE remain unclear, this article aims to provide a framework for conceptualizing across all ages and highlight the implications they may have for longevity.
Collapse
Affiliation(s)
- Adrian Flores
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, USA
- Department of Cellular and Integrative Physiology, UNMC, Omaha, NE, USA
| | - Nghi M. Nguyen
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, USA
- Department of Genetics, Cell Biology and Anatomy, UNMC, Omaha, NE, USA
| | - Gurudutt Pendyala
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, USA
- Department of Genetics, Cell Biology and Anatomy, UNMC, Omaha, NE, USA
- Child Health Research Institute, Omaha, NE, USA
- National Strategic Research Institute, UNMC, Omaha, NE, USA
| |
Collapse
|
9
|
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: 3] [Impact Index Per Article: 1.5] [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
|
10
|
Lepore G, Morley-McLaughlin T, Davidson N, Han C, Masese C, Reynolds G, Saltz V, Robinson SA. Buprenorphine reduces somatic withdrawal in a mouse model of early-life morphine exposure. Drug Alcohol Depend 2023; 248:109938. [PMID: 37267743 DOI: 10.1016/j.drugalcdep.2023.109938] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 06/04/2023]
Abstract
The rising prevalence of early-life opioid exposure has become a pressing public health issue in the U.S. Neonates exposed to opioids in utero are at risk of experiencing a constellation of postpartum withdrawal symptoms commonly referred to as neonatal opioid withdrawal syndrome (NOWS). Buprenorphine (BPN), a partial agonist at the mu-opioid receptor (MOR) and antagonist at the kappa-opioid receptor (KOR), is currently approved to treat opioid use disorder in adult populations. Recent research suggests that BPN may also be effective in reducing withdrawal symptoms in neonates who were exposed to opioids in utero. We sought to determine whether BPN attenuates somatic withdrawal in a mouse model of NOWS. Our findings indicate that the administration of morphine (10mg/kg, s.c.) from postnatal day (PND) 1-14 results in increased somatic symptoms upon naloxone-precipitated (1mg/kg, s.c.) withdrawal. Co-administration of BPN (0.3mg/kg, s.c.) from PND 12-14 attenuated symptoms in morphine-treated mice. On PND 15, 24h following naloxone-precipitated withdrawal, a subset of mice was examined for thermal sensitivity in the hot plate test. BPN treatment significantly increased response latency in morphine-exposed mice. Lastly, neonatal morphine exposure elevated mRNA expression of KOR, and reduced mRNA expression of corticotropin-releasing hormone (CRH) in the periaqueductal gray when measured on PND 14. Altogether, this data provides support for the therapeutic effects of acute low-dose buprenorphine treatment in a mouse model of neonatal opioid exposure and withdrawal.
Collapse
Affiliation(s)
- Gina Lepore
- Department of Systems Pharmacology and Translational Therapeutics. Perelman School of Medicine, University of PennsylvaniaPhiladelphiaPA19104, United States
| | | | - Natalie Davidson
- Department of Psychology, Williams CollegeWilliamsMA01267, United States
| | - Caitlin Han
- Department of Psychology, Williams CollegeWilliamsMA01267, United States
| | - Cynthia Masese
- Department of Psychology, Williams CollegeWilliamsMA01267, United States
| | - Grace Reynolds
- Department of Psychology, Williams CollegeWilliamsMA01267, United States
| | - Victoria Saltz
- Department of Psychology, Williams CollegeWilliamsMA01267, United States
| | - Shivon A Robinson
- Department of Psychology, Williams CollegeWilliamsMA01267, United States.
| |
Collapse
|
11
|
Hornburg KJ, Slosky LM, Cofer G, Cook J, Qi Y, Porkka F, Clark NB, Pires A, Petrella JR, White LE, Wetsel WC, Barak L, Caron MG, Johnson GA. Prenatal heroin exposure alters brain morphology and connectivity in adolescent mice. NMR IN BIOMEDICINE 2023; 36:e4842. [PMID: 36259728 PMCID: PMC10483958 DOI: 10.1002/nbm.4842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/30/2022] [Accepted: 10/05/2022] [Indexed: 06/16/2023]
Abstract
The United States is experiencing a dramatic increase in maternal opioid misuse and, consequently, the number of individuals exposed to opioids in utero. Prenatal opioid exposure has both acute and long-lasting effects on health and wellbeing. Effects on the brain, often identified at school age, manifest as cognitive impairment, attention deficit, and reduced scholastic achievement. The neurobiological basis for these effects is poorly understood. Here, we examine how in utero exposure to heroin affects brain development into early adolescence in a mouse model. Pregnant C57BL/6J mice received escalating doses of heroin twice daily on gestational days 4-18. The brains of offspring were assessed on postnatal day 28 using 9.4 T diffusion MRI of postmortem specimens at 36 μm resolution. Whole-brain volumes and the volumes of 166 bilateral regions were compared between heroin-exposed and control offspring. We identified a reduction in whole-brain volume in heroin-exposed offspring and heroin-associated volume changes in 29 regions after standardizing for whole-brain volume. Regions with bilaterally reduced standardized volumes in heroin-exposed offspring relative to controls include the ectorhinal and insular cortices. Regions with bilaterally increased standardized volumes in heroin-exposed offspring relative to controls include the periaqueductal gray, septal region, striatum, and hypothalamus. Leveraging microscopic resolution diffusion tensor imaging and precise regional parcellation, we generated whole-brain structural MRI diffusion connectomes. Using a dimension reduction approach with multivariate analysis of variance to assess group differences in the connectome, we found that in utero heroin exposure altered structure-based connectivity of the left septal region and the region that acts as a hub for limbic regulatory actions. Consistent with clinical evidence, our findings suggest that prenatal opioid exposure may have effects on brain morphology, connectivity, and, consequently, function that persist into adolescence. This work expands our understanding of the risks associated with opioid misuse during pregnancy and identifies biomarkers that may facilitate diagnosis and treatment.
Collapse
Affiliation(s)
- Kathryn J. Hornburg
- Department of Radiology, School of Medicine, Duke University; 311 Research Drive; Campus Box 3302; Durham, NC 27710 United States
| | - Lauren M. Slosky
- Department of Cell Biology, School of Medicine, Duke University; Campus Box 3709; Durham, NC 27710 United States
- Department of Pharmacology, University of Minnesota; 312 Church Street SE; 3-104 Nils Hasselmo Hall; Minneapolis, MN 55455 United States
| | - Gary Cofer
- Department of Radiology, School of Medicine, Duke University; 311 Research Drive; Campus Box 3302; Durham, NC 27710 United States
| | - James Cook
- Department of Radiology, School of Medicine, Duke University; 311 Research Drive; Campus Box 3302; Durham, NC 27710 United States
| | - Yi Qi
- Department of Radiology, School of Medicine, Duke University; 311 Research Drive; Campus Box 3302; Durham, NC 27710 United States
| | - Fiona Porkka
- Department of Cell Biology, School of Medicine, Duke University; Campus Box 3709; Durham, NC 27710 United States
| | - Nicholas B. Clark
- Department of Cell Biology, School of Medicine, Duke University; Campus Box 3709; Durham, NC 27710 United States
| | - Andrea Pires
- Department of Cell Biology, School of Medicine, Duke University; Campus Box 3709; Durham, NC 27710 United States
| | - Jeffrey R Petrella
- Department of Radiology, School of Medicine, Duke University; 311 Research Drive; Campus Box 3302; Durham, NC 27710 United States
| | - Leonard E. White
- Department of Neurology, School of Medicine, Duke University; Campus Box 2900; Durham, NC 27710 United States
| | - William C. Wetsel
- Department of Cell Biology, School of Medicine, Duke University; Campus Box 3709; Durham, NC 27710 United States
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Duke University; Campus Box 102508; Durham, NC 27710 United States
- Department of Neurology, School of Medicine, Duke University; Campus Box 2900; Durham, NC 27710 United States
| | - Lawrence Barak
- Department of Cell Biology, School of Medicine, Duke University; Campus Box 3709; Durham, NC 27710 United States
| | - Marc G. Caron
- Department of Cell Biology, School of Medicine, Duke University; Campus Box 3709; Durham, NC 27710 United States
- Department of Neurology, School of Medicine, Duke University; Campus Box 2900; Durham, NC 27710 United States
| | - G. Allan Johnson
- Department of Radiology, School of Medicine, Duke University; 311 Research Drive; Campus Box 3302; Durham, NC 27710 United States
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University; Campus Box 90281; Durham, NC 27708-0281 United States
| |
Collapse
|
12
|
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
|
13
|
Elam HB, Donegan JJ, Hsieh J, Lodge DJ. Gestational buprenorphine exposure disrupts dopamine neuron activity and related behaviors in adulthood. eNeuro 2022; 9:ENEURO.0499-21.2022. [PMID: 35851301 PMCID: PMC9337603 DOI: 10.1523/eneuro.0499-21.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 05/13/2022] [Accepted: 05/20/2022] [Indexed: 11/21/2022] Open
Abstract
Opioid misuse among pregnant women is rapidly increasing in the United States. The number of maternal opioid-related diagnoses increased by 131% in the last ten years, resulting in an increased number of infants exposed to opioids in utero and a subsequent increase in infants developing neonatal abstinence syndrome (NAS). The most prescribed treatment to combat maternal opioid use disorder is buprenorphine, a partial μ-opioid receptor agonist and κ-opioid receptor antagonist. Buprenorphine treatment effectively reduces NAS but has been associated with disrupted cortical development and neurodevelopmental consequences in childhood. Less is known about the long-term neurodevelopmental consequences following buprenorphine exposure in utero Previous research has shown that gestational buprenorphine exposure can induce anxiety- and depressive-like phenotypes in adult rats, suggesting that exposure to buprenorphine in utero may render individuals more susceptible to psychiatric illness in adulthood. A common pathology observed across multiple psychiatric illnesses is dopamine system dysfunction. Here, we administered the highly-abused opioid, oxycodone (10 mg/kg, i.p.) or a therapeutic used to treat opioid use disorder, buprenorphine (1 mg/kg, i.p) to pregnant Sprague Dawley rats from gestational day 11 through 21, then examined neurophysiological alterations in the mesolimbic dopamine system and dopamine-dependent behaviors in adult offspring. We found that gestational exposure to buprenorphine or oxycodone increases dopamine neuron activity in adulthood. Moreover, prenatal buprenorphine exposure disrupts the afferent regulation of dopamine neuron activity in the ventral tegmental area (VTA). Taken together, we posit that gestational buprenorphine or oxycodone exposure can have profound effects on the mesolimbic dopamine system in adulthood.Significance StatementThe opioid epidemic in the United States is a growing problem that affects people from all demographics, including pregnant women. In 2017, nearly 21,000 pregnant women reported misusing opioids during pregnancy, which can lead to many physiological and neurodevelopmental complications in infants. To combat illicit opioid use during pregnancy, buprenorphine is the priority treatment option, as it reduces illicit opioid use and alleviates symptoms of neonatal abstinence syndrome in infants. However, less is known about the long-term neurophysiological consequences of in utero opioid or buprenorphine exposure. Here, we demonstrate that both oxycodone and buprenorphine exposure, in utero, can result in aberrant dopamine system function in adult rats. These results provide evidence of potential long-lasting effects of opioid exposure during development.
Collapse
Affiliation(s)
- Hannah B Elam
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Jennifer J Donegan
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, 78229, USA
- Department of Psychiatry and Behavioral Sciences, Dell Medical School at UT Austin, Austin, TX, USA
| | - Jenny Hsieh
- Department of Neuroscience, Developmental and Regenerative Biology, University of Texas at San Antonio, San Antonio, TX, 78249, USA
- Brain Health Consortium, University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Daniel J Lodge
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, 78229, USA
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, USA
| |
Collapse
|
14
|
Allen MC, Moog NK, Buss C, Yen E, Gustafsson HC, Sullivan EL, Graham AM. Co-occurrence of preconception maternal childhood adversity and opioid use during pregnancy: Implications for offspring brain development. Neurotoxicol Teratol 2021; 88:107033. [PMID: 34601061 PMCID: PMC8578395 DOI: 10.1016/j.ntt.2021.107033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 09/20/2021] [Accepted: 09/24/2021] [Indexed: 12/11/2022]
Abstract
Understanding of the effects of in utero opioid exposure on neurodevelopment is a priority given the recent dramatic increase in opioid use among pregnant individuals. However, opioid abuse does not occur in isolation-pregnant individuals abusing opioids often have a significant history of adverse experiences in childhood, among other co-occurring factors. Understanding the specific pathways in which these frequently co-occurring factors may interact and cumulatively influence offspring brain development in utero represents a priority for future research in this area. We highlight maternal history of childhood adversity (CA) as one such co-occurring factor that is more prevalent among individuals using opioids during pregnancy and which is increasingly shown to affect offspring neurodevelopment through mechanisms beginning in utero. Despite the high incidence of CA history in pregnant individuals using opioids, we understand very little about the effects of comorbid prenatal opioid exposure and maternal CA history on fetal brain development. Here, we first provide an overview of current knowledge regarding effects of opioid exposure and maternal CA on offspring neurodevelopment that may occur during gestation. We then outline potential mechanistic pathways through which these factors might have interactive and cumulative influences on offspring neurodevelopment as a foundation for future research in this area.
Collapse
Affiliation(s)
- Madeleine C Allen
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States
| | - Nora K Moog
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Psychology, Luisenstrasse 57, 10117 Berlin, Germany
| | - Claudia Buss
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Psychology, Luisenstrasse 57, 10117 Berlin, Germany; Development, Health and Disease Research Program, University of California, Irvine, 837 Health Sciences Drive, Irvine, California 92697, United States
| | - Elizabeth Yen
- Department of Pediatrics, Tufts Medical Center, Boston, MA 02111, United States
| | - Hanna C Gustafsson
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States
| | - Elinor L Sullivan
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States; Division of Neuroscience, Oregon National Primate Research Center, 505 NW 185(th) Ave., Beaverton, OR 97006, United States; Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States
| | - Alice M Graham
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States.
| |
Collapse
|
15
|
Vassoler FM, Wimmer ME. Consequences of Parental Opioid Exposure on Neurophysiology, Behavior, and Health in the Next Generations. Cold Spring Harb Perspect Med 2021; 11:a040436. [PMID: 32601130 PMCID: PMC8485740 DOI: 10.1101/cshperspect.a040436] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Substance abuse and the ongoing opioid epidemic represents a large societal burden. This review will consider the long-term impact of opioid exposure on future generations. Prenatal, perinatal, and preconception exposure are reviewed with discussion of both maternal and paternal influences. Opioid exposure can have long-lasting effects on reproductive function, gametogenesis, and germline epigenetic programming, which can influence embryogenesis and alter the developmental trajectory of progeny. The potential mechanisms by which preconception maternal and paternal opioid exposure produce deleterious consequences on the health, behavior, and physiology of offspring that have been identified by clinical and animal studies will be discussed. The timing, nature, dosing, and duration of prenatal opioid exposure combined with other important environmental considerations influence the extent to which these manipulations affect parents and their progeny. Epigenetic inheritance refers to the transmission of environmental insults across generations via mechanisms independent of the DNA sequence. This topic will be further explored in the context of prenatal, perinatal, and preconception opioid exposure for both the maternal and paternal lineage.
Collapse
Affiliation(s)
- Fair M Vassoler
- Tufts University, Cummings School of Veterinary Medicine, Grafton, Massachusetts 01536, USA
| | - Mathieu E Wimmer
- Department of Psychology and Program in Neuroscience, Temple University, Philadelphia, Pennsylvania 19122, USA
| |
Collapse
|
16
|
Minakova E, Sarafinovska S, Mikati MO, Barclay KM, McCullough KB, Dougherty JD, Al-Hasani R, Maloney SE. Ontogenetic Oxycodone Exposure Affects Early Life Communicative Behaviors, Sensorimotor Reflexes, and Weight Trajectory in Mice. Front Behav Neurosci 2021; 15:615798. [PMID: 33692675 PMCID: PMC7937712 DOI: 10.3389/fnbeh.2021.615798] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/29/2021] [Indexed: 12/21/2022] Open
Abstract
Nationwide, opioid misuse among pregnant women has risen four-fold from 1999 to 2014, with commensurate increase in neonates hospitalized for neonatal abstinence syndrome (NAS). NAS occurs when a fetus exposed to opioids in utero goes into rapid withdrawal after birth. NAS treatment via continued post-natal opioid exposure has been suggested to worsen neurodevelopmental outcomes. We developed a novel model to characterize the impact of in utero and prolonged post-natal oxycodone (Oxy) exposure on early behavior and development. Via subcutaneous pump implanted before breeding, C57BL/6J dams were infused with Oxy at 10 mg/kg/day from conception through pup-weaning. At birth, in utero oxy-exposed pups were either cross-fostered (paired with non-Oxy exposed dams) to model opioid abstinence (in utero Oxy) or reared by their biological dams still receiving Oxy to model continued post-natal opioid exposure (prolonged Oxy). Offspring from vehicle-exposed dams served as cross-fostered (in utero Veh) or biologically reared (prolonged Veh) controls. In utero Oxy exposure resulted in sex-dependent weight reductions and altered spectrotemporal features of isolation-induced ultrasonic vocalization (USV). Meanwhile, prolonged Oxy pups exhibited reduced weight and sex-differential delays in righting reflex. Specifically, prolonged Oxy female offspring exhibited increased latency to righting. Prolonged Oxy pups also showed decreases in number of USV calls and changes to spectrotemporal USV features. Overall, ontogenetic Oxy exposure was associated with impaired attainment of gross and sensorimotor milestones, as well as alterations in communication and affective behaviors, indicating a need for therapeutic interventions. The model developed here will enable studies of withdrawal physiology and opioid-mediated mechanisms underlying these neurodevelopmental deficits.
Collapse
Affiliation(s)
- Elena Minakova
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, United States
| | - Simona Sarafinovska
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, United States
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, United States
- Medical Scientist Training Program, Washington University in St. Louis, St. Louis, MO, United States
| | - Marwa O. Mikati
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, United States
- Division of Biology and Biomedical Sciences, Washington University in St. Louis, St. Louis, MO, United States
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States
- Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, United States
- Center for Clinical Pharmacology, St. Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis, MO, United States
| | - Kia M. Barclay
- Division of Biology and Biomedical Sciences, Washington University in St. Louis, St. Louis, MO, United States
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States
- Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, United States
- Center for Clinical Pharmacology, St. Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis, MO, United States
| | - Katherine B. McCullough
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, United States
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, United States
| | - Joseph D. Dougherty
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, United States
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, United States
- Intellectual and Developmental Disabilities Research Center, Washington University In St. Louis, St. Louis, MO, United States
| | - Ream Al-Hasani
- Division of Biology and Biomedical Sciences, Washington University in St. Louis, St. Louis, MO, United States
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States
- Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, United States
- Center for Clinical Pharmacology, St. Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis, MO, United States
| | - Susan E. Maloney
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, United States
- Intellectual and Developmental Disabilities Research Center, Washington University In St. Louis, St. Louis, MO, United States
| |
Collapse
|
17
|
Sun CY, Li JR, Wang YY, Lin SY, Ou YC, Lin CJ, Wang JD, Liao SL, Chen CJ. Indoxyl sulfate caused behavioral abnormality and neurodegeneration in mice with unilateral nephrectomy. Aging (Albany NY) 2021; 13:6681-6701. [PMID: 33621199 PMCID: PMC7993681 DOI: 10.18632/aging.202523] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 12/29/2020] [Indexed: 12/17/2022]
Abstract
Chronic Kidney Disease (CKD) and neurodegenerative diseases are aging-related diseases. CKD with declined renal function is associated with an elevation of circulating indoxyl sulfate, a metabolite synthesized by gut microbes. We explored the roles of gut microbial metabolites in linking with Central Nervous System (CNS) diseases by administrating indoxyl sulfate intraperitoneally to male C57BL/6 mice with unilateral nephrectomy. Upon exposure, the accumulation of indoxyl sulfate was noted in the blood, prefrontal cortical tissues, and cerebrospinal fluid. Mice showed behavioral signs of mood disorders and neurodegeneration such as anxiety, depression, and cognitive impairment. Those behavioral changes were accompanied by disturbed neuronal survival, neural stem cell activity, expression of Brain-Derived Neurotrophic Factor, serotonin, corticosterone, and Repressor Element-1 Silencing Transcription Factor, and post-receptor intracellular signaling, as well as upregulated oxidative stress and neuroinflammation. Uremic toxin adsorbent AST-120 improved the above mentioned changes. Intriguingly, intracerebroventricular indoxyl sulfate administration only caused limited alterations in the normal mice and the alterations were reversed by aryl hydrocarbon receptor antagonism. The findings suggest pathogenic roles of indoxyl sulfate in the development of CNS diseases, and highlight gut microbiota as alternative targets for intervention with the aim of slowing down the progression of CKD and decreasing CNS complications.
Collapse
Affiliation(s)
- Chiao-Yin Sun
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung 204, Taiwan
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung 204, Taiwan
- Kidney Research Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- School of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Jian-Ri Li
- Division of Urology, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Ya-Yu Wang
- Department of Family Medicine, Taichung Veterans General Hospital, Taichung 407, Taiwan
- Institute of Clinical Medicine, National Yang Ming University, Taipei 112, Taiwan
| | - Shih-Yi Lin
- Institute of Clinical Medicine, National Yang Ming University, Taipei 112, Taiwan
- Center for Geriatrics and Gerontology, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Yen-Chuan Ou
- Department of Urology, Tungs’ Taichung MetroHarbor Hospital, Taichung 435, Taiwan
| | - Cheng-Jui Lin
- Division of Nephrology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei 104, Taiwan
- Mackay Junior College of Medicine, Nursing and Management, Taipei 251, Taiwan
| | - Jiaan-Der Wang
- Children’s Medical Center, Taichung Veterans General Hospital, Taichung 407, Taiwan
- Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung 407, Taiwan
| | - Su-Lan Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404, Taiwan
- Ph.D. Program in Translational Medicine, College of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan
| |
Collapse
|
18
|
Liu QF, Park SW, Kim YM, Song SJ, Chin YW, Pak SC, Jeon S, Koo BS. Administration of Kyung-Ok-Ko reduces stress-induced depressive behaviors in mice through inhibition of inflammation pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 265:113441. [PMID: 33027642 DOI: 10.1016/j.jep.2020.113441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 09/24/2020] [Accepted: 09/29/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Kyung-Ok-Ko (KOK), a traditional medicinal formula composed of Rehmannia glutinosa (Gaertn.) DC, Poria cocos (Schw.) Wolf, Korean Red Panax ginseng C.A.Mey, and honey, has been used to treat amnesia and dementia. KOK has also been shown to ameliorate transient cerebral global ischemia-induced brain damage, but the antidepressant-like effect of KOK has not been examined. AIM OF THE STUDY This study examined the antidepressant-like effect of KOK in an immobilization-induced stress mouse and its mechanisms of action. MATERIALS AND METHODS The animals in the stress group were immobilized for two hours a day for two weeks. KOK at a dose of 1 g/kg/day was administered orally to the stressed mice for two weeks in advance of their immobilization. A forced swimming test was performed to analyze their depressive behaviors. To examine the anti-inflammatory or antioxidative effects of KOK, the murine macrophage cell line, RAW 264.7 cells and human neuroblastoma cell, SH-SY5Y cells, were treated with lipopolysaccharide (LPS) and hydrogen peroxide, respectively. RESULT The KOK extract showed no significant toxicity when the cells were treated with a KOK extract at 5, 10, 25, 50, and 100 μg/mL. The KOK ethanol extract reduced LPS-induced TNF-α production, inducible nitric oxide (iNOS) mRNA level, and the levels of MAPK and p38 phosphorylation in RAW 264.7 cells. KOK also suppressed H2O2-induced cell death and the production of reactive oxygen species (ROS) in SH-SY5Y cells. In the forced swimming test, KOK induced a decrease in immobility and an increase in climbing activity. Finally, the administration of KOK reversed the up-regulation of IkB-α phosphorylation in the stressed mouse cortex. CONCLUSION KOK might be useful for the treatment of depression caused by environmental and lifestyle-related stress.
Collapse
Affiliation(s)
- Quan Feng Liu
- Department of Oriental Neuropsychiatry, Graduate School of Oriental Medicine, Dongguk University, 814 Siksa-dong, Goyang-si, Gyeonggi-do, 10326, Republic of Korea
| | - Sun-Woo Park
- Yaksan Korean Medical Clinic 302, Dongyang-plaza 533-3, Yatap-dong, Bundang-gu, Seongnam-si, Gyeonggi-do, South Korea
| | - Young-Mi Kim
- College of Pharmacy, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Sue-Jin Song
- Department of Oriental Neuropsychiatry, Graduate School of Oriental Medicine, Dongguk University, 814 Siksa-dong, Goyang-si, Gyeonggi-do, 10326, Republic of Korea
| | - Young-Won Chin
- College of Pharmacy, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Sok Cheon Pak
- School of Biomedical Sciences, Charles Sturt University, Bathurst, NSW, 2795, Australia
| | - Songhee Jeon
- Department of Biomedical Sciences, Center for Creative Biomedical Scientists at Chonnam National University, Gwangju, 61469, Republic of Korea.
| | - Byung-Soo Koo
- Department of Oriental Neuropsychiatry, Graduate School of Oriental Medicine, Dongguk University, 814 Siksa-dong, Goyang-si, Gyeonggi-do, 10326, Republic of Korea; Department of Korean Neuropsychiatry, Dongguk University Ilsan Hospital, 32, Dongguk-lo, Ilsandong-gu, Goyang, Gyeonggi-do, 10326, Republic of Korea.
| |
Collapse
|
19
|
Sun CY, Li JR, Wang YY, Lin SY, Ou YC, Lin CJ, Wang JD, Liao SL, Chen CJ. p-Cresol Sulfate Caused Behavior Disorders and Neurodegeneration in Mice with Unilateral Nephrectomy Involving Oxidative Stress and Neuroinflammation. Int J Mol Sci 2020; 21:6687. [PMID: 32932690 PMCID: PMC7555291 DOI: 10.3390/ijms21186687] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/05/2020] [Accepted: 09/10/2020] [Indexed: 12/15/2022] Open
Abstract
Protein-bound uremic toxins, such as p-cresol sulfate (PCS), can be accumulated with declined renal function and aging and is closely linked with central nervous system (CNS) diseases. In the periphery, PCS has effects on oxidative stress and inflammation. Since oxidative stress and inflammation have substantial roles in the pathogenesis of neurological disorders, the CNS effects of PCS were investigated in unilateral nephrectomized C57/BL/6 mice. Unlike intact mice, unilateral nephrectomized mice showed increased circulating levels of PCS after exogenous administration. Upon PCS exposure, the unilateral nephrectomized mice developed depression-like, anxiety-like, and cognitive impairment behaviors with brain PCS accumulation in comparison with the nephrectomy-only group. In the prefrontal cortical tissues, neuronal cell survival and neurogenesis were impaired along with increased apoptosis, oxidative stress, and neuroinflammation. Circulating brain-derived neurotrophic factors (BDNF) and serotonin were decreased in association with increased corticosterone and repressor element-1 silencing transcription factor (REST), regulators involved in neurological disorders. On the contrary, these PCS-induced changes were alleviated by uremic toxin absorbent AST-120. Taken together, PCS administration in mice with nephrectomy contributed to neurological disorders with increased oxidative stress and neuroinflammation, which were alleviated by PCS chelation. It is suggested that PCS may be a therapeutic target for chronic kidney disease-associated CNS diseases.
Collapse
Affiliation(s)
- Chiao-Yin Sun
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung 20401, Taiwan;
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
- Kidney Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- School of Medicine, Chang Gung University, Taoyuan 33305, Taiwan
| | - Jian-Ri Li
- Division of Urology, Taichung Veterans General Hospital, Taichung 40705, Taiwan;
| | - Ya-Yu Wang
- Department of Family Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan;
- Institute of Clinical Medicine, National Yang Ming University, Taipei 112304, Taiwan;
| | - Shih-Yi Lin
- Institute of Clinical Medicine, National Yang Ming University, Taipei 112304, Taiwan;
- Center for Geriatrics and Gerontology, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| | - Yen-Chuan Ou
- Department of Urology, Tungs’ Taichung MetroHarbor Hospital, Taichung 43304, Taiwan;
| | - Cheng-Jui Lin
- Division of Nephrology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei 10449, Taiwan;
- Mackay Junior College of Medicine, Nursing and Mangement, Taipei 11260, Taiwan
| | - Jiaan-Der Wang
- Children’s Medical Center, Taichung Veterans General Hospital, Taichung 40705, Taiwan;
- Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung 407224, Taiwan
| | - Su-Lan Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40705, Taiwan;
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40705, Taiwan;
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 40402, Taiwan
- Program in Translational Medicine, College of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan
| |
Collapse
|
20
|
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: 15] [Impact Index Per Article: 3.0] [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
|
21
|
Prenatal exposure to methadone or buprenorphine impairs cognitive performance in young adult rats. Drug Alcohol Depend 2020; 212:108008. [PMID: 32402939 DOI: 10.1016/j.drugalcdep.2020.108008] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/20/2020] [Accepted: 03/30/2020] [Indexed: 11/23/2022]
Abstract
BACKGROUND Concerns have been raised about the use of opioid maintenance treatment (OMT) during pregnancy and negative effects for the offspring. While neonatal outcomes and short-term effects are relatively well described, studies examining long-term effects in adolescents and adults are absent. The aim of the present study was to examine effects on learning and memory in young adult rats prenatally exposed to methadone or buprenorphine. METHODS Female rats were implanted with a 28-day osmotic minipump delivering methadone (10 mg/kg/day), buprenorphine (1 mg/kg/day) or vehicle 5 days prior to mating. To examine possible effects on cognitive functioning, young adult offspring were included in three different behavioral tests that examine recognition memory, nonspatial, and spatial learning and memory. In addition, offspring growth and maternal behavior after birh were investigated. RESULTS Prenatal exposure to methadone or buprenorphine caused impaired recognition memory and nonspatial reference learning and memory in young adult rats compared with the vehicle-treated group. Methadone-exposed offspring, but not the buprenorphine-exposed, also showed reduced long-term spatial memory. We did not observe any changes in maternal behavior or offspring growth after prenatal exposure to methadone or buprenorphine, suggesting that the impaired cognitive functioning is due to the opioid exposure rather than reduced maternal caregiving. CONCLUSION The present findings of long-term cognitive impairments in methadone- and buprenorphine-exposed offspring points to a negative impact of OMT on neurobiological development.
Collapse
|
22
|
Boggess T, Risher WC. Clinical and basic research investigations into the long-term effects of prenatal opioid exposure on brain development. J Neurosci Res 2020; 100:396-409. [PMID: 32459039 DOI: 10.1002/jnr.24642] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/01/2020] [Accepted: 04/27/2020] [Indexed: 12/26/2022]
Abstract
Coincident with the opioid epidemic in the United States has been a dramatic increase in the number of children born with neonatal abstinence syndrome (NAS), a form of withdrawal resulting from opioid exposure during pregnancy. Many research efforts on NAS have focused on short-term care, including acute symptom treatment and weaning of the infants off their drug dependency prior to authorizing their release. However, investigations into the long-term effects of prenatal opioid exposure (POE) on brain development, from the cellular to the behavioral level, have not been as frequent. Given the importance of the perinatal period for human brain development, opioid-induced disturbances in the formation and function of nascent synaptic networks and glia have the potential to impact brain connectivity and cognition long after the drug supply is cutoff shortly after birth. In this review, we will summarize the current state of NAS research, bringing together findings from human studies and preclinical animal models to highlight what is known about how POE can induce significant, prolonged deficits in brain structure and function. With rates of NAS continuing to rise, particularly in regions that already face substantial socioeconomic challenges, we speculate as to the most promising avenues for future research to alleviate this growing multigenerational threat.
Collapse
Affiliation(s)
- Taylor Boggess
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV, USA
| | - W Christopher Risher
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV, USA
| |
Collapse
|
23
|
Abstract
BACKGROUND Depression and post-traumatic stress disorder (PTSD) are leading causes of disability and loss of life by suicide. Currently, there are less than satisfactory medical solutions to treat these mental disorders. Here, we explore recent preclinical and clinical studies demonstrating the potential of using buprenorphine to treat major depressive disorder, treatment-resistant depression, and PTSD. METHOD Bibliographic databases were searched to include preclinical and clinical studies demonstrating the therapeutic potential of buprenorphine and the involvement of the kappa opioid receptor (KOR) in mediating these effects. RESULTS Original clinical studies examining the effectiveness of buprenorphine to treat depression were mixed. The majority of participants in the PTSD studies were males and suffer from chronic pain and/or substance use disorders. Nonetheless, these recent studies and analyses established proof of concept warranting farther investigations. Additionally, KOR likely mediates the antidepressant and some of the anxiolytic effects of buprenorphine. Still, it appears that the full spectrum of buprenorphine's beneficial effects might be due to activity at other opioid receptors as well. CONCLUSIONS Pharmaceuticals' abilities to treat medical conditions directly relates to their ability to act upon the endogenous biological systems related to the conditions. Thus, these recent findings are likely a reflection of the central role that the endogenous opioid system has in these mental illnesses. Further studies are necessary to study the involvement of endogenous opioid systems, and specifically KOR, in mediating buprenorphine's beneficial effects and the ability to treat these medical conditions while minimizing risks for misuse and diversion.
Collapse
Affiliation(s)
- Caitlin A Madison
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX77843, USA
| | - Shoshana Eitan
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX77843, USA
| |
Collapse
|
24
|
Wallin CM, Bowen SE, Roberge CL, Richardson LM, Brummelte S. Gestational buprenorphine exposure: Effects on pregnancy, development, neonatal opioid withdrawal syndrome, and behavior in a translational rodent model. Drug Alcohol Depend 2019; 205:107625. [PMID: 31706250 DOI: 10.1016/j.drugalcdep.2019.107625] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/27/2019] [Accepted: 09/16/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND The opioid crisis has led to an increased number of pregnant opioid-dependent women receiving opioid-maintenance therapy (e.g. buprenorphine, BUP), but little is known about the consequences of gestational BUP exposure on pregnancy outcomes, maternal care, or offspring development. METHODS Our translational rodent model began BUP exposure to adult female rats (N = 30) at least 7 days before conception and continued throughout the postpartum period. Both therapeutic low-dose (BUP-LD, 0.3 mg/kg, s.c.) and overexposure high-dose (BUP-HD, 1.0 mg/kg) doses of BUP were compared to saline control. Female rats were bred in house with drug-naïve adult male rats. The day after parturition, litters were culled to 5 males/5 females and assigned randomly to various behavioral tests and assessed either neonates or adolescents. Litter characteristics, maternal caregiving, Neonatal Opioid Withdrawal Syndrome (NOWS), offspring development and adolescent behaviors were evaluated. RESULTS BUP-LD decreased maternal care, delayed offspring development, decreased offspring body weight, length, temperature, and pain sensitivity (p's < .05). BUP-HD drastically reduced maternal care and offspring survival, altered litter characteristics, and increased NOWS (p's < .05). CONCLUSION These results demonstrate that the therapeutic BUP-LD in rats was relatively safe with subtle effects on maternal care and rodent offspring. However, overexposure BUP-HD in rats produced NOWS and compromised maternal caregiving as well as rodent offspring survival. More research is critical to validate the translational implication of these findings for human opioid-dependent mothers maintained on BUP-maintenance therapy.
Collapse
Affiliation(s)
- Chela M Wallin
- Department of Psychology, Wayne State University, Detroit, MI, 48202, USA.
| | - Scott E Bowen
- Department of Psychology, Wayne State University, Detroit, MI, 48202, USA.
| | - Chelsea L Roberge
- Department of Psychology, Wayne State University, Detroit, MI, 48202, USA.
| | | | - Susanne Brummelte
- Department of Psychology, Wayne State University, Detroit, MI, 48202, USA.
| |
Collapse
|
25
|
Gao Q, Shi Y, Liao M, Xiao J, Li X, Zhou L, Liu C, Liu P, Cao H. Laboratory and field evaluation of the aphidicidal activity of moso bamboo (Phyllostachys pubescens) leaf extract and identification of the active components. PEST MANAGEMENT SCIENCE 2019; 75:3167-3174. [PMID: 30941856 DOI: 10.1002/ps.5434] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/27/2019] [Accepted: 03/30/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Botanical pesticides increasingly play important roles in the control of agricultural pests. In this study, the aphidicidal effect of moso bamboo (Phyllostachys pubescens) extract against mustard aphid was confirmed, the main active compounds identified, and aphidicidal mechanism of the most active compound established. RESULTS When the treatment concentration was 10.0 g L-1 , the corrected mortality of bamboo leaf extract (BE) was 53.22 ± 5.20% and the petroleum ether component of bamboo leaf extract (PE) reached 82.76 ± 4.50%, which also showed a synergistic effect with imidacloprid. Four flavonoids were identified as the main active components in the BE via activity tracking and phytochemical method. Isoorientin had an LC50 of 313.22 mg L-1 , and affected the activities of acetylcholinesterase and peroxidase significantly, revealing the possible aphidicidal mechanism. When the treatment of 11.1% PE·imidacloprid was 200 mL, the control effect was 99.07%, which was better than that observed with 10% of imidacloprid or 0.5% of matrine. CONCLUSIONS These data provide a better understanding of the aphidicidal activity and aphidicidal mechanism of moso bamboo leaf extract and the most active compound, isoorientin. This will help in developing a more effective botanical aphicide. © 2019 Society of Chemical Industry.
Collapse
Affiliation(s)
- Quan Gao
- School of Plant Protection, Anhui Agricultural University, Hefei, China
| | - Yanhong Shi
- Provincial Key Laboratory for Agri-Food Safety, Hefei, China
- School of Resource and Environment, Anhui Agricultural University, Hefei, China
| | - Min Liao
- School of Plant Protection, Anhui Agricultural University, Hefei, China
| | - Jinjing Xiao
- School of Plant Protection, Anhui Agricultural University, Hefei, China
| | - Xiuxia Li
- School of Plant Protection, Anhui Agricultural University, Hefei, China
| | - Lijun Zhou
- School of Plant Protection, Anhui Agricultural University, Hefei, China
| | - Chengwu Liu
- Provincial Key Laboratory for Agri-Food Safety, Hefei, China
| | - Peng Liu
- Provincial Key Laboratory for Agri-Food Safety, Hefei, China
| | - Haiqun Cao
- School of Plant Protection, Anhui Agricultural University, Hefei, China
- Provincial Key Laboratory for Agri-Food Safety, Hefei, China
| |
Collapse
|
26
|
Kongstorp M, Bogen IL, Stiris T, Andersen JM. High Accumulation of Methadone Compared with Buprenorphine in Fetal Rat Brain after Maternal Exposure. J Pharmacol Exp Ther 2019; 371:130-137. [PMID: 31358559 DOI: 10.1124/jpet.119.259531] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/18/2019] [Indexed: 12/12/2022] Open
Abstract
Experimental animal studies are valuable in revealing a causal relationship between prenatal exposure to opioid maintenance treatment (OMT) and subsequent effects; however, previous animal studies of OMT during pregnancy have been criticized for their lack of clinical relevance because of their use of high drug doses and the absence of pharmacokinetic data. Hence, the aim of this study was to determine blood and brain concentrations in rat dams, fetuses, and offspring after continuous maternal exposure to methadone or buprenorphine during gestation and to examine the offspring for neonatal outcomes and withdrawal symptoms. Female rats were implanted with a 28-day osmotic minipump delivering methadone (10 mg/kg per day), buprenorphine (1 mg/kg per day) or vehicle 5 days before mating. Continuous exposure to methadone or buprenorphine induced stable blood concentrations in the dams of 0.25 ± 0.02 µM and 5.65 ± 0.16 nM, respectively. The fetal brain concentration of methadone (1.89 ± 0.35 nmol/g) was twice as high as that in the maternal brain, whereas the fetal brain concentration of buprenorphine (20.02 ± 4.97 pmol/g) was one-third the maternal brain concentration. The opioids remained in the offspring brain several days after the exposure ceased. Offspring prenatally exposed to methadone, but not buprenorphine, displayed reduced body weight and length and increased corticosterone levels. No significant changes in ultrasonic vocalizations were revealed. Our data in rat fetuses and neonates indicate that OMT with buprenorphine may be a better choice than methadone during pregnancy. SIGNIFICANCE STATEMENT: Concern has been raised about the use of opioid maintenance treatment during pregnancy because of the important role of the endogenous opioid system in brain development. Here, we show that the methadone concentration in the fetal rat brain was twice as high as that in the maternal brain, whereas the buprenorphine concentration was one-third the maternal concentration. Furthermore, buprenorphine allowed more favorable birth outcomes, suggesting that buprenorphine may be a better choice during pregnancy.
Collapse
Affiliation(s)
- Mette Kongstorp
- Section for Drug Abuse Research, Department of Forensic Sciences (M.K., I.L.B., J.M.A.) and Department of Neonatal Intensive Care (T.S.), Oslo University Hospital, and Institute of Clinical Medicine, Faculty of Medicine (M.K., T.S.), Department of Pharmacy, Faculty of Mathematics and Natural Sciences (J.M.A.), and Institute of Basic Medical Sciences, Faculty of Medicine (I.L.B.), University of Oslo, Oslo, Norway
| | - Inger Lise Bogen
- Section for Drug Abuse Research, Department of Forensic Sciences (M.K., I.L.B., J.M.A.) and Department of Neonatal Intensive Care (T.S.), Oslo University Hospital, and Institute of Clinical Medicine, Faculty of Medicine (M.K., T.S.), Department of Pharmacy, Faculty of Mathematics and Natural Sciences (J.M.A.), and Institute of Basic Medical Sciences, Faculty of Medicine (I.L.B.), University of Oslo, Oslo, Norway
| | - Tom Stiris
- Section for Drug Abuse Research, Department of Forensic Sciences (M.K., I.L.B., J.M.A.) and Department of Neonatal Intensive Care (T.S.), Oslo University Hospital, and Institute of Clinical Medicine, Faculty of Medicine (M.K., T.S.), Department of Pharmacy, Faculty of Mathematics and Natural Sciences (J.M.A.), and Institute of Basic Medical Sciences, Faculty of Medicine (I.L.B.), University of Oslo, Oslo, Norway
| | - Jannike Mørch Andersen
- Section for Drug Abuse Research, Department of Forensic Sciences (M.K., I.L.B., J.M.A.) and Department of Neonatal Intensive Care (T.S.), Oslo University Hospital, and Institute of Clinical Medicine, Faculty of Medicine (M.K., T.S.), Department of Pharmacy, Faculty of Mathematics and Natural Sciences (J.M.A.), and Institute of Basic Medical Sciences, Faculty of Medicine (I.L.B.), University of Oslo, Oslo, Norway
| |
Collapse
|
27
|
Swain JE, Ho SS, Fox H, Garry D, Brummelte S. Effects of opioids on the parental brain in health and disease. Front Neuroendocrinol 2019; 54:100766. [PMID: 31128130 PMCID: PMC8318357 DOI: 10.1016/j.yfrne.2019.100766] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 12/20/2022]
Abstract
The epidemic of opioid use disorder (OUD) directly affects millions of women of child-bearing age. Unfortunately, parenting behaviors - among the most important processes for human survival - are vulnerable to the effects of OUD. The standard of care for pregnant women with OUD is opioid maintenance therapy (OMT), of which the primary objective is to mitigate addiction-related stress. The aim of this review is to synthesize current information specific to pregnancy and parenting that may be affected by OUD. We first summarize a model of the parental brain supported by animal research and human neuroimaging. We then review animal models of exogenous opioid effects on parental brain and behavior. We also present preliminary data for a unifying hypothesis that may link different effects of exogenous opioids on parenting across species and in the context of OMT. Finally, we discuss future directions that may inform research and clinical decision making for peripartum women with OUD.
Collapse
Affiliation(s)
- James E Swain
- Department of Psychiatry and Behavioral Health, and Psychology, Stony Brook University, Stony Brook, NY, United States; Department of Psychiatry, Psychology, and Center for Human Growth & Development, University of Michigan, Ann Arbor, MI, United States.
| | - S Shaun Ho
- Department of Psychiatry and Behavioral Health, and Psychology, Stony Brook University, Stony Brook, NY, United States
| | - Helen Fox
- Department of Psychiatry and Behavioral Health, and Psychology, Stony Brook University, Stony Brook, NY, United States
| | - David Garry
- Department of Obstetrics and Gynecology, Stony Brook University, Stony Brook, NY, United States
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, Detroit, MI, United States.
| |
Collapse
|
28
|
Mortensen NP, Caffaro MM, Snyder RW, Yueh YL, Fennell TR. Placental trophoblast transfer of opioids following exposures to individual or mixtures of opioids in vitro. Exp Biol Med (Maywood) 2019; 244:846-849. [PMID: 31091988 DOI: 10.1177/1535370219851109] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Infants born with neonatal abstinence syndrome increased 5-fold between 2000 and 2012. Prenatal exposure to opioids has been linked to altered brain development, congenital heart defects, neural tube defects, and clubfoot. In the study presented here, placental transfer rate of six opioids; morphine, heroin, fentanyl, methadone, oxycodone, and naloxone was compared in vitro using a human trophoblast cell monolayer model, BeWo b30. The rate and concentration of opioid translocation were investigated individually and in mixtures. The opioid transfer was quantified at 5, 15, 30, 60, and 120 min, using target liquid chromatography-mass spectrometry. The transfer was lowest for heroin (<1.2% of administered dose at 120 min; the permeability across the cells [P]: 0.21 × 10−5 cm/s) and highest for oxycodone (13.2% of administered dose at 120 min; P: 2.46 × 10−5 cm/s). As oxycodone is a preferred drug for both short- and long-term pain treatment, more knowledge is needed to understand the potential adverse impact on fetal development, growth, and well-being. Opioid exposure is likely to happen as mixtures, and we therefore investigated the transfer of selected mixtures. Permeability was significantly decreased for heroin when administered together with fentanyl. This study demonstrates that the transfer rate between individual opioids varies significantly and that some mixtures may impact the transfer of some individual opioids like heroin. Impact statement Gaining knowledge about opioid exposure and placental transfer of opioids is an important part of providing clinicians with the necessary information for therapeutic strategies during pregnancy in the rising opioid epidemic. Opioids exposure often includes a mixture rather than an exposure to an individual opioid. We investigated placental trophoblast permeability to opioids in vitro for six individual opioids and selected mixtures. The rate and concentration of opioid translocation across placental trophoblast monolayer varied considerably between the six investigated opioids. Oxycodone had the highest level of translocation. We also found that the transfer of heroin decreased when administered together with fentanyl, while no change was observed for fentanyl transfer in the mixture. This suggests that opioid mixtures may decrease translocation of some opioids while others are unaffected.
Collapse
Affiliation(s)
| | | | | | - Yun L Yueh
- Discovery Sciences, RTI International, RTP NC 27709, USA
| | | |
Collapse
|
29
|
Tsai SY, Bendriem RM, Lee CTD. The cellular basis of fetal endoplasmic reticulum stress and oxidative stress in drug-induced neurodevelopmental deficits. Neurobiol Stress 2019; 10:100145. [PMID: 30937351 PMCID: PMC6430408 DOI: 10.1016/j.ynstr.2018.100145] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 12/02/2018] [Accepted: 12/26/2018] [Indexed: 11/30/2022] Open
Abstract
Prenatal substance exposure is a growing public health concern worldwide. Although the opioid crisis remains one of the most prevalent addiction problems in our society, abuse of cocaine, methamphetamines, and other illicit drugs, particularly amongst pregnant women, are nonetheless significant and widespread. Evidence demonstrates prenatal drug exposure can affect fetal brain development and thus can have long-lasting impact on neurobehavioral and cognitive performance later in life. In this review, we highlight research examining the most prevalent drugs of abuse and their effects on brain development with a focus on endoplasmic reticulum stress and oxidative stress signaling pathways. A thorough exploration of drug-induced cellular stress mechanisms during prenatal brain development may provide insight into therapeutic interventions to combat effects of prenatal drug exposure.
Collapse
Affiliation(s)
- S-Y.A. Tsai
- Integrative Neuroscience Branch, Division of Neuroscience and Behavior, National Institute on Drug Abuse, The National Institute of Health, Department of Health and Human Services, Bethesda, MD, 20892, USA
| | - Raphael M. Bendriem
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Chun-Ting D. Lee
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, USA
| |
Collapse
|
30
|
Byrnes EM, Vassoler FM. Modeling prenatal opioid exposure in animals: Current findings and future directions. Front Neuroendocrinol 2018; 51:1-13. [PMID: 28965857 PMCID: PMC5649358 DOI: 10.1016/j.yfrne.2017.09.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 09/26/2017] [Accepted: 09/27/2017] [Indexed: 01/03/2023]
Abstract
The past decade has seen a drastic rise in the number of infants exposed to opioids in utero. It is unclear what lasting effect this exposure may have on these children. Animal models of prenatal opioid exposure may provide insight into potential areas of vulnerability. The present review summarizes the findings across animal models of prenatal opioid exposure, including exposure to morphine, methadone, buprenorphine, and oxycodone. Details regarding the drug, doses, and duration of treatment, as well as key findings, are summarized in tables with associated references. Finally, significant gaps in the current preclinical literature and future directions are discussed.
Collapse
Affiliation(s)
- Elizabeth M Byrnes
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, United States.
| | - Fair M Vassoler
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, United States
| |
Collapse
|
31
|
Gilardi F, Augsburger M, Thomas A. Will Widespread Synthetic Opioid Consumption Induce Epigenetic Consequences in Future Generations? Front Pharmacol 2018; 9:702. [PMID: 30018553 PMCID: PMC6037745 DOI: 10.3389/fphar.2018.00702] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/11/2018] [Indexed: 11/13/2022] Open
Abstract
A growing number of evidence demonstrates that ancestral exposure to xenobiotics (pollutants, drugs of abuse, etc.) can perturb the physiology and behavior of descendants. Both maternal and paternal transmission of phenotype across generations has been proved, demonstrating that parental drug history may have significant implications for subsequent generations. In the last years, the burden of novel synthetic opioid (NSO) consumption, due to increased medical prescription of pain medications and to easier accessibility of these substances on illegal market, is raising new questions first in term of public health, but also about the consequences of the parental use of these drugs on future generations. Besides being associated to the neonatal abstinence syndrome, in utero exposure to opioids has an impact on neuronal development with long-term repercussions that are potentially transmitted to subsequent generations. In addition, recent reports suggest that opioid use even before conception influences the reactivity to opioids of the progeny and the following generations, likely through epigenetic mechanisms. This review describes the current knowledge about the transgenerational effects of opioid consumption. We summarize the preclinical and clinical findings showing the implications for the subsequent generations of parental exposure to opioids earlier in life. Limitations of the existing data on NSOs and new perspectives of the research are also discussed, as well as clinical and forensic consequences.
Collapse
Affiliation(s)
- Federica Gilardi
- Forensic Toxicology and Chemistry Unit, University Center of Legal Medicine, Lausanne University Hospital - Geneva University Hospitals, Geneva, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Marc Augsburger
- Forensic Toxicology and Chemistry Unit, University Center of Legal Medicine, Lausanne University Hospital - Geneva University Hospitals, Geneva, Switzerland
| | - Aurelien Thomas
- Forensic Toxicology and Chemistry Unit, University Center of Legal Medicine, Lausanne University Hospital - Geneva University Hospitals, Geneva, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
32
|
Wu CC, Hung CJ, Lin SY, Wang YY, Chang CY, Chen WY, Liao SL, Raung SL, Yang CP, Chen CJ. Treadmill exercise alleviated prenatal buprenorphine exposure-induced depression in rats. Neurochem Int 2017; 110:91-100. [PMID: 28962922 DOI: 10.1016/j.neuint.2017.09.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/05/2017] [Accepted: 09/24/2017] [Indexed: 01/16/2023]
Abstract
Mounting evidence suggests that physical exercise shows health benefits in a range of diseases, including psychiatric disorders. Perinatal opioid exposure produces neurobehavioral abnormality, which includes depression symptoms, in patients and their offspring following chronic use of buprenorphine, a mixed agonist/antagonist with a high affinity to opioid receptors, for pain control. Previously, we demonstrated that prenatal buprenorphine exposure in pregnant Sprague-Dawley rats starting from gestation day 7 and lasting for 14 days caused the development of depression-like phenotypes in pups at postnatal day 21. Using the same prenatal buprenorphine exposure model, we further demonstrated that a 4-week course of moderate treadmill exercise conducted on pups starting from postnatal day 22 improved depression-like neurobehaviors. Prenatal buprenorphine exposure-induced neurobehavioral changes were accompanied by reductions of neuronal survival, neural stem cell-associated genes, plasma level of brain-derived neurotrophic factor (BDNF) and serotonin, phosphorylated tropomyosin-related kinase receptor type B (TrkB), phosphorylated extracellular signal-regulated kinase (ERK), PKA activity, phosphorylated cAMP response element-binding protein (CREB), and CREB DNA binding activity, as well as elevation of repressor element-1 silencing transcription factor (REST), oxidative stress, and inflammatory responses. Those changes in parameters of plasma and brain were improved by treadmill exercise. In conclusion, the findings of the current study suggest that a non-pharmacological option, i.e., moderate treadmill exercise, alleviated the development of depression-like neurobehaviors by resolving the oxidative and inflammatory burden as well as by enhancing neurochemical and neuroendocrine signaling.
Collapse
Affiliation(s)
- Chih-Cheng Wu
- Department of Anesthesiology, Taichung Veterans General Hospital, Taichung City 407, Taiwan; Department of Financial and Computational Mathematics, Providence University, Taichung City 433, Taiwan
| | - Chih-Jen Hung
- Department of Anesthesiology, Taichung Veterans General Hospital, Taichung City 407, Taiwan
| | - Shih-Yi Lin
- Center for Geriatrics and Gerontology, Taichung Veterans General Hospital, Taichung City 407, Taiwan
| | - Ya-Yu Wang
- Division of Family Medicine, Taichung Veterans General Hospital, Taichung City 407, Taiwan
| | - Cheng-Yi Chang
- Department of Surgery, Feng Yuan Hospital, Taichung City 420, Taiwan
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung Hsing University, Taichung City 402, Taiwan
| | - Su-Lan Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City 407, Taiwan
| | - Shue-Ling Raung
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City 407, Taiwan
| | - Ching-Ping Yang
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City 407, Taiwan
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City 407, Taiwan; Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung City 404, Taiwan.
| |
Collapse
|
33
|
Cui R, Fan J, Ge T, Tang L, Li B. The mechanism of acute fasting-induced antidepressant-like effects in mice. J Cell Mol Med 2017; 22:223-229. [PMID: 28782175 PMCID: PMC5742683 DOI: 10.1111/jcmm.13310] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/19/2017] [Indexed: 11/30/2022] Open
Abstract
Acute fasting induced antidepressant‐like effects. However, the exact brain region and mechanism of these actions are still largely unknown. Therefore, in this study the antidepressant‐like effects of acute fasting on c‐Fos expression and BDNF levels were investigated. Consistent with our previous findings, immobility time was remarkably shortened by 9 hrs fasting in the forced swimming test. Furthermore, these antidepressant‐like effects of 9 fasting were inhibited by a 5‐HT2A/2C receptor agonist (±)‐1‐(2, 5‐dimethoxy‐4‐iodophenyl)‐2‐aminopropane hydrochloride (DOI), and the effect of DOI was blocked by pretreatment with a selective 5‐HT2A receptor antagonist ketanserin. Immunohistochemical study has shown that c‐Fos level was significantly increased by 9 hrs fasting in prefrontal cortex but not hippocampus and habenular. Fasting‐induced c‐Fos expression was further enhanced by DOI in prefrontal cortex, and these enhancements were inhibited by ketanserin. The increased BDNF levels by fasting were markedly inhibited by DOI in frontal cortex and hippocampus, and these effects of DOI on BDNF levels were also blocked by ketanserin. These findings suggest that the antidepressant‐like effects of acute fasting may be exerted via 5‐HT2A receptor and particularly sensitive to neural activity in the prefrontal cortex. Furthermore, these antidepressant‐like effects are also mediated by CREB and BDNF pathway in hippocampus and frontal cortex. Therefore, fasting may be potentially helpful against depression.
Collapse
Affiliation(s)
- Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Jie Fan
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Tongtong Ge
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Linda Tang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
34
|
Tran TH, Griffin BL, Stone RH, Vest KM, Todd TJ. Methadone, Buprenorphine, and Naltrexone for the Treatment of Opioid Use Disorder in Pregnant Women. Pharmacotherapy 2017; 37:824-839. [PMID: 28543191 DOI: 10.1002/phar.1958] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Pregnant women with opioid use disorder can be treated with methadone, buprenorphine, or naltrexone to reduce opioid use and improve retention to treatment. In this review, we compare the pregnancy outcomes of methadone, buprenorphine, and naltrexone in clinical trials and discuss the potential behavioral and developmental effects of these agents seen in offspring in animal studies. Important clinical considerations in the management of opioid use disorder in pregnant women and their infants are also discussed. Outside of pregnancy, buprenorphine is used in combination with naloxone to reduce opioid abuse and diversion. During pregnancy, however, the use of buprenorphine as a single agent is preferred to prevent prenatal naloxone exposure. Both methadone and buprenorphine are widely used to treat opioid use disorder; however, compared with methadone, buprenorphine is associated with shorter treatment duration, less medication needed to treat neonatal abstinence syndrome (NAS) symptoms, and shorter hospitalizations for neonates. Despite being the standard of care, medication-assisted treatment with methadone or buprenorphine is still underused, making it apparent that more options are necessary. Naltrexone is not a first-line treatment primarily because both detoxification and an opioid-free period are required. More research is needed to determine naltrexone safety and benefits in pregnant women. Animal studies suggest that changes in pain sensitivity, developmental processes, and behavioral responses may occur in children born to mothers receiving methadone, buprenorphine, or naltrexone and is an area that warrants future studies.
Collapse
Affiliation(s)
- Tran H Tran
- Pharmacy Practice, Midwestern University Chicago College of Pharmacy, Downers Grove, Illinois
| | - Brooke L Griffin
- Pharmacy Practice, Midwestern University Chicago College of Pharmacy, Downers Grove, Illinois
| | - Rebecca H Stone
- Pharmacy Practice, University of Georgia College of Pharmacy, Athens, Georgia
| | - Kathleen M Vest
- Pharmacy Practice, Midwestern University Chicago College of Pharmacy, Downers Grove, Illinois
| | - Timothy J Todd
- Pharmacy Practice, Midwestern University Chicago College of Pharmacy, Downers Grove, Illinois
| |
Collapse
|
35
|
Hauser KF, Knapp PE. Opiate Drugs with Abuse Liability Hijack the Endogenous Opioid System to Disrupt Neuronal and Glial Maturation in the Central Nervous System. Front Pediatr 2017; 5:294. [PMID: 29410949 PMCID: PMC5787058 DOI: 10.3389/fped.2017.00294] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 12/20/2017] [Indexed: 01/19/2023] Open
Abstract
The endogenous opioid system, comprised of multiple opioid neuropeptide and receptor gene families, is highly expressed by developing neural cells and can significantly influence neuronal and glial maturation. In many central nervous system (CNS) regions, the expression of opioid peptides and receptors occurs only transiently during development, effectively disappearing with subsequent maturation only to reemerge under pathologic conditions, such as with inflammation or injury. Opiate drugs with abuse liability act to modify growth and development by mimicking the actions of endogenous opioids. Although typically mediated by μ-opioid receptors, opiate drugs can also act through δ- and κ-opioid receptors to modulate growth in a cell-type, region-specific, and developmentally regulated manner. Opioids act as biological response modifiers and their actions are highly contextual, plastic, modifiable, and influenced by other physiological processes or pathophysiological conditions, such as neuro-acquired immunodeficiency syndrome. To date, most studies have considered the acute effects of opiates on cellular maturation. For example, activating opioid receptors typically results in acute growth inhibition in both neurons and glia. However, with sustained opioid exposure, compensatory factors become operative, a concept that has been largely overlooked during CNS maturation. Accordingly, this article surveys prior studies on the effects of opiates on CNS maturation, and also suggests new directions for future research in this area. Identifying the cellular and molecular mechanisms underlying the adaptive responses to chronic opiate exposure (e.g., tolerance) during maturation is crucial toward understanding the consequences of perinatal opiate exposure on the CNS.
Collapse
Affiliation(s)
- Kurt F Hauser
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States.,Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States.,Institute for Drug and Alcohol Studies, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Pamela E Knapp
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States.,Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States.,Institute for Drug and Alcohol Studies, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| |
Collapse
|
36
|
Kidder IJ, Mudery JA, Barreda S, Taska DJ, Bailey EF. Evaluating the control: minipump implantation and breathing behavior in the neonatal rat. J Appl Physiol (1985) 2016; 121:615-22. [PMID: 27402557 PMCID: PMC11735011 DOI: 10.1152/japplphysiol.00080.2016] [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: 01/27/2016] [Accepted: 07/05/2016] [Indexed: 11/22/2022] Open
Abstract
We evaluated genioglossus (GG) gross motoneuron morphology, electromyographic (EMG) activities, and respiratory patterning in rat pups allowed to develop without interference (unexposed) and pups born to dams subjected to osmotic minipump implantation in utero (saline-exposed). In experiment 1, 48 Sprague-Dawley rat pups (Charles-River Laboratories), ages postnatal day 7 (P7) through postnatal day 10 (P10), were drawn from two experimental groups, saline-exposed (n = 24) and unexposed (n = 24), and studied on P7, P8, P9, or P10. Pups in both groups were sedated (Inactin hydrate, 70 mg/kg), and fine-wire electrodes were inserted into the GG muscle of the tongue and intercostal muscles to record EMG activities during breathing in air and at three levels of normoxic hypercapnia [inspired CO2 fraction (FiCO2 ): 0.03, 0.06, and 0.09]. Using this approach, we assessed breathing frequency, heart rate, apnea type, respiratory event types, and respiratory stability. In experiment 2, 16 rat pups were drawn from the same experimental groups, saline-exposed (n = 9) and unexposed (n = 7), and used in motoneuron-labeling studies. In these pups a retrograde dye was injected into the GG muscle, and the brain stems were subsequently harvested and sliced. Labeled GG motoneurons were identified with microscopy, impaled, and filled with Lucifer yellow. Double-labeled motoneurons were reconstructed, and the number of primary projections and soma volumes were calculated. Whereas pups in each group exhibited the same number (P = 0.226) and duration (P = 0.093) of respiratory event types and comparable motoneuron morphologies, pups in the implant group exhibited more central apneas and respiratory instability relative to pups allowed to develop without interference.
Collapse
Affiliation(s)
- Ian J Kidder
- Department of Physiology, College of Medicine, The University of Arizona, Tucson, Arizona; and
| | - Jordan A Mudery
- Department of Physiology, College of Medicine, The University of Arizona, Tucson, Arizona; and
| | - Santiago Barreda
- Department of Linguistics, University of California Davis, Davis, California
| | - David J Taska
- Department of Physiology, College of Medicine, The University of Arizona, Tucson, Arizona; and
| | - E Fiona Bailey
- Department of Physiology, College of Medicine, The University of Arizona, Tucson, Arizona; and
| |
Collapse
|
37
|
Fitting S, Zou S, El-Hage N, Suzuki M, Paris JJ, Schier CJ, Rodríguez JW, Rodriguez M, Knapp PE, Hauser KF. Opiate addiction therapies and HIV-1 Tat: interactive effects on glial [Ca²⁺]i, oxyradical and neuroinflammatory chemokine production and correlative neurotoxicity. Curr HIV Res 2015; 12:424-34. [PMID: 25760046 PMCID: PMC4475822 DOI: 10.2174/1570162x1206150311161147] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 12/13/2014] [Accepted: 02/12/2015] [Indexed: 01/05/2023]
Abstract
Few preclinical studies have compared the relative therapeutic efficacy of medications used to treat opiate addiction in relation to neuroAIDS. Here we compare the ability of methadone and buprenorphine, and the prototypic opiate morphine, to potentiate the neurotoxic and proinflammatory ([Ca2+]i, ROS, H2O2, chemokines) effects of HIV-1 Tat in neuronal and/or mixed-glial co-cultures. Repeated observations of neurons during 48 h exposure to combinations of Tat, equimolar concentrations (500 nM) of morphine, methadone, or buprenorphine exacerbated neurotoxicity significantly above levels seen with Tat alone. Buprenorphine alone displayed marked neurotoxicity at 500 nM, prompting additional studies of its neurotoxic effects at 5 nM and 50 nM concentrations ± Tat. In combination with Tat, buprenorphine displayed paradoxical, concentration-dependent, neurotoxic and neuroprotective actions. Buprenorphine neurotoxicity coincided with marked elevations in [Ca2+]i, but not increases in glial ROS or chemokine release. Tat by itself elevated the production of CCL5/RANTES, CCL4/MIP-1β, and CCL2/MCP-1. Methadone and buprenorphine alone had no effect, but methadone interacted with Tat to further increase production of CCL5/RANTES. In combination with Tat, all drugs significantly increased glial [Ca2+]i, but ROS was only significantly increased by co-exposure with morphine. Taken together, the increases in glial [Ca2+]i, ROS, and neuroinflammatory chemokines were not especially accurate predictors of neurotoxicity. Despite similarities, opiates displayed differences in their neurotoxic and neuroinflammatory interactions with Tat. Buprenorphine, in particular, was partially neuroprotective at a low concentration, which may result from its unique pharmacological profile at multiple opioid receptors. Overall, the results reveal differences among addiction medications that may impact neuroAIDS.
Collapse
|
38
|
Abstract
This paper is the thirty-sixth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2013 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior, and the roles of these opioid peptides and receptors in pain and analgesia; stress and social status; tolerance and dependence; learning and memory; eating and drinking; alcohol and drugs of abuse; sexual activity and hormones, pregnancy, development and endocrinology; mental illness and mood; seizures and neurologic disorders; electrical-related activity and neurophysiology; general activity and locomotion; gastrointestinal, renal and hepatic functions; cardiovascular responses; respiration and thermoregulation; and immunological responses.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
39
|
Jeon S, Lee CH, Liu QF, Kim GW, Koo BS, Pak SC. Alteration in brain-derived neurotrophic factor (BDNF) after treatment of mice with herbal mixture containing Euphoria longana, Houttuynia cordata and Dioscorea japonica. ACTA ACUST UNITED AC 2014; 22:77. [PMID: 25431319 PMCID: PMC4268823 DOI: 10.1186/s40199-014-0077-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 11/14/2014] [Indexed: 01/20/2023]
Abstract
BACKGROUND Literature data indicate that brain-derived neurotrophic factor (BDNF), cyclic-AMP response element-binding protein (CREB) and phospho-CREB (pCREB) may have a place in depression. BDNF belongs to the neurotrophin family that plays an important role in proliferation, survival and differentiation of different cell populations in the mammalian nervous system. The herbal mixture used in the present study consists of Euphoria longana, Houttuynia cordata and Dioscorea japonica. The purpose of the present study was to determine the neuroprotective effect of herbal mixture. We also tested the hypothesis that administration of herbs reverses memory deficits and promotes the protein expression of BDNF in the mouse brain. METHODS Mice were randomized into four different treatment groups (n = 10/group). Normal and stress groups received regular lab chow without stress and under stress conditions, respectively, for 3 weeks. The animals in the stress group were immobilized for 4 hours a day for 2 weeks. Different doses of herbal mixture (206 and 618 mg/kg) were administered for 3 weeks to those mice under stress conditions. Mice were analyzed by behavioral tests and immunoblotting examination in the hippocampus and cortex. An additional in vitro investigation was performed to examine whether herbs induce neurotoxicity in a human neuroblastoma cell line, SH-SY5Y cells. RESULTS No significant toxicity of herbs on human neuroblastoma cells was observed. These herbs demonstrated an inductive effect on the expression of BDNF, pCREB and pAkt. For spatial working memory test, herbal mixture fed mice exhibited an increased level of spontaneous alternation (p < 0.01) compared to those in stress conditions. Moreover, herbal mixture produced highly significant (p < 0.01) reduction in the immobility time in the tail suspension test. Mice in the herbal mixture groups demonstrated lower serum corticosterone concentration than mice in the stress group (p < 0.05). Effects of the oral administration of herbal mixture on protein levels of BDNF in the hippocampi and cortices were significant. CONCLUSIONS Our study showed that herbal mixture administration has antidepressant effects in mice. It is proposed that adverse events such as stress and depression can modulate the expression of molecular players of cellular plasticity in the brain.
Collapse
Affiliation(s)
- Songhee Jeon
- Dongguk University Research Institute of Biotechnology, Seoul, 100-715, Republic of Korea.
| | - Chia-Hung Lee
- Department of Neuropsychiatry, Graduate School of Oriental Medicine, Dongguk University, Gyeongju, Republic of Korea.
| | - Quan Feng Liu
- Department of Neuropsychiatry, Graduate School of Oriental Medicine, Dongguk University, Gyeongju, Republic of Korea.
| | - Geun Woo Kim
- Department of Korean Neuropsychiatry, Dongguk University Bundang Oriental Hospital, Sungnam, Republic of Korea.
| | - Byung-Soo Koo
- Department of Korean Neuropsychiatry, Dongguk University Ilsan Oriental Hospital, Goyang, Republic of Korea.
| | - Sok Cheon Pak
- School of Biomedical Sciences, Charles Sturt University, Bathurst, NSW, 2795, Australia.
| |
Collapse
|
40
|
Rodent models of depression: neurotrophic and neuroinflammatory biomarkers. BIOMED RESEARCH INTERNATIONAL 2014; 2014:932757. [PMID: 24999483 PMCID: PMC4066721 DOI: 10.1155/2014/932757] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 05/18/2014] [Indexed: 12/13/2022]
Abstract
Rodent models are an indispensable tool for studying etiology and progress of depression. Since interrelated systems of neurotrophic factors and cytokines comprise major regulatory mechanisms controlling normal brain plasticity, impairments of these systems form the basis for development of cerebral pathologies, including mental diseases. The present review focuses on the numerous experimental rodent models of depression induced by different stress factors (exteroceptive and interoceptive) during early life (including prenatal period) or adulthood, giving emphasis to the data on the changes of neurotrophic factors and neuroinflammatory indices in the brain. These parameters are closely related to behavioral depression-like symptoms and impairments of neuronal plasticity and are both gender- and genotype-dependent. Stress-related changes in expression of neurotrophins and cytokines in rodent brain are region-specific. Some contradictory data reported by different groups may be a consequence of differences of stress paradigms or their realization in different laboratories. Like all experimental models, stress-induced depression-like conditions are experimental simplification of clinical depression states; however, they are suitable for understanding the involvement of neurotrophic factors and cytokines in the pathogenesis of the disease—a goal unachievable in the clinical reality. These major regulatory systems may be important targets for therapeutic measures as well as for development of drugs for treatment of depression states.
Collapse
|