1
|
Drury KM, Hall TA, Orwoll B, Adhikary S, Kirby A, Williams CN. Exposure to Sedation and Analgesia Medications: Short-term Cognitive Outcomes in Pediatric Critical Care Survivors With Acquired Brain Injury. J Intensive Care Med 2024; 39:374-386. [PMID: 37885235 PMCID: PMC11132562 DOI: 10.1177/08850666231210261] [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] [Indexed: 10/28/2023]
Abstract
Background/Objective: Pediatric intensive care unit (PICU) survivors risk significant cognitive morbidity, particularly those with acquired brain injury (ABI) diagnoses. Studies show sedative and analgesic medication may potentiate neurologic injury, but few studies evaluate impact on survivor outcomes. This study aimed to evaluate whether exposures to analgesic and sedative medications are associated with worse neurocognitive outcome. Methods: A retrospective cohort study was conducted of 91 patients aged 8 to 18 years, undergoing clinical neurocognitive evaluation approximately 1 to 3 months after PICU discharge. Electronic health data was queried for sedative and analgesic medication exposures, including opioids, benzodiazepines, propofol, ketamine, and dexmedetomidine. Doses were converted to class equivalents, evaluated by any exposure and cumulative dose exposure per patient weight. Cognitive outcome was derived from 8 objective cognitive assessments with an emphasis on executive function skills using Principal Components Analysis. Then, linear regression was used to control for baseline cognitive function estimates to calculate a standardized residualized neurocognitive index (rNCI) z-score. Multivariable linear regression evaluated the association between rNCI and medication exposure controlling for covariates. Significance was defined as P < .05. Results: Most (n = 80; 88%) patients received 1 or more study medications. Any exposure and higher cumulative doses of benzodiazepine and ketamine were significantly associated with worse rNCI in bivariate analyses. When controlling for Medicaid, preadmission comorbid conditions, length of stay, delirium, and receipt of other medication classes, receipt of benzodiazepine was associated with significantly worse rNCI (β-coefficient = -0.48, 95% confidence interval = -0.88, -0.08). Conclusions: Exposure to benzodiazepines was independently associated with worse acute phase cognitive outcome using objective assessments focused on executive function skills when controlling for demographic and illness characteristics. Clinician decisions regarding medication regimens in the PICU may serve as a modifiable factor to improve outcomes. Additional inquiry into associations with long-term cognitive outcome and optimal medication regimens is needed.
Collapse
Affiliation(s)
- Kurt M. Drury
- Department of Pediatrics, Division of Critical Care, Oregon Health & Science University
- Pediatric Critical Care and Neurotrauma Recovery Program, Oregon Health & Science University
| | - Trevor A. Hall
- Pediatric Critical Care and Neurotrauma Recovery Program, Oregon Health & Science University
- Department of Pediatrics, Division of Pediatric Psychology, Oregon Health & Science University
| | - Benjamin Orwoll
- Department of Pediatrics, Division of Critical Care, Oregon Health & Science University
| | - Sweta Adhikary
- Pediatric Critical Care and Neurotrauma Recovery Program, Oregon Health & Science University
- School of Medicine, Oregon Health and Science University
| | - Aileen Kirby
- Department of Pediatrics, Division of Critical Care, Oregon Health & Science University
| | - Cydni N. Williams
- Department of Pediatrics, Division of Critical Care, Oregon Health & Science University
- Pediatric Critical Care and Neurotrauma Recovery Program, Oregon Health & Science University
| |
Collapse
|
2
|
Cha J, Filatov G, Smith SJ, Gammaitoni AR, Lothe A, Reeder T. Fenfluramine increases survival and reduces markers of neurodegeneration in a mouse model of Dravet syndrome. Epilepsia Open 2024; 9:300-313. [PMID: 38018342 PMCID: PMC10839300 DOI: 10.1002/epi4.12873] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 11/21/2023] [Indexed: 11/30/2023] Open
Abstract
OBJECTIVE In patients with Dravet syndrome (DS), fenfluramine reduced convulsive seizure frequency and provided clinical benefit in nonseizure endpoints (e.g., executive function, survival). In zebrafish mutant scn1 DS models, chronic fenfluramine treatment preserved neuronal cytoarchitecture prior to seizure onset and prevented gliosis; here, we extend these findings to a mammalian model of DS (Scn1a+/- mice) by evaluating the effects of fenfluramine on neuroinflammation (degenerated myelin, activated microglia) and survival. METHODS Scn1a+/- DS mice were treated subcutaneously once daily with fenfluramine (15 mg/kg) or vehicle from postnatal day (PND) 7 until 35-37. Sagittal brain sections were processed for immunohistochemistry using antibodies to degraded myelin basic protein (D-MBP) for degenerated myelin, or CD11b for activated (inflammatory) microglia; sections were scored semi-quantitatively. Apoptotic nuclei were quantified by TUNEL assay. Statistical significance was evaluated by 1-way ANOVA with post-hoc Dunnett's test (D-MBP, CD11b, and TUNEL) or Logrank Mantel-Cox (survival). RESULTS Quantitation of D-MBP immunostaining per 0.1 mm2 unit area of the parietal cortex and hippocampus CA3 yielded significantly higher spheroidal and punctate myelin debris counts in vehicle-treated DS mice than in wild-type mice. Fenfluramine treatment in DS mice significantly reduced these counts. Activated CD11b + microglia were more abundant in DS mouse corpus callosum and hippocampus than in wild-type controls. Fenfluramine treatment of DS mice resulted in significantly fewer activated CD11b + microglia than vehicle-treated DS mice in these brain regions. TUNEL staining in corpus callosum was increased in DS mice relative to wild-type controls. Fenfluramine treatment in DS mice lowered TUNEL staining relative to vehicle-treated DS mice. By PND 35-37, 55% of control DS mice had died, compared with 24% of DS mice receiving fenfluramine treatment (P = 0.0291). SIGNIFICANCE This is the first report of anti-neuroinflammation and pro-survival after fenfluramine treatment in a mammalian DS model. These results corroborate prior data in humans and animal models and suggest important pharmacological activities for fenfluramine beyond seizure reduction. PLAIN LANGUAGE SUMMARY Dravet syndrome is a severe epilepsy disorder that impairs learning and causes premature death. Clinical studies in patients with Dravet syndrome show that fenfluramine reduces convulsive seizures. Additional studies suggest that fenfluramine may have benefits beyond seizures, including promoting survival and improving control over emotions and behavior. Our study is the first to use a Dravet mouse model to investigate nonseizure outcomes of fenfluramine. Results showed that fenfluramine treatment of Dravet mice reduced neuroinflammation significantly more than saline treatment. Fenfluramine-treated Dravet mice also lived longer than saline-treated mice. These results support clinical observations that fenfluramine may have benefits beyond seizures.
Collapse
Affiliation(s)
- John Cha
- University of California San FranciscoSan FranciscoCaliforniaUSA
- Zogenix, Inc. (now a part of UCB)EmeryvilleCaliforniaUSA
| | - Gregory Filatov
- Zogenix, Inc. (now a part of UCB)EmeryvilleCaliforniaUSA
- Crosshair Therapeutics, Inc.SunnyvaleCaliforniaUSA
| | - Steven J. Smith
- Zogenix, Inc. (now a part of UCB)EmeryvilleCaliforniaUSA
- WuXi AppTec, Inc.San FranciscoCaliforniaUSA
| | | | | | - Thadd Reeder
- Zogenix, Inc. (now a part of UCB)EmeryvilleCaliforniaUSA
| |
Collapse
|
3
|
Useinovic N, Near M, Cabrera OH, Boscolo A, Milosevic A, Harvey R, Newson A, Chastain-Potts S, Quillinan N, Jevtovic-Todorovic V. Neonatal sevoflurane exposure induces long-term changes in dendritic morphology in juvenile rats and mice. Exp Biol Med (Maywood) 2023; 248:641-655. [PMID: 37309741 PMCID: PMC10350807 DOI: 10.1177/15353702231170003] [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: 02/10/2023] [Accepted: 03/11/2023] [Indexed: 06/14/2023] Open
Abstract
General anesthetics are potent neurotoxins when given during early development, causing apoptotic deletion of substantial number of neurons and persistent neurocognitive and behavioral deficits in animals and humans. The period of intense synaptogenesis coincides with the peak of susceptibility to deleterious effects of anesthetics, a phenomenon particularly pronounced in vulnerable brain regions such as subiculum. With steadily accumulating evidence confirming that clinical doses and durations of anesthetics may permanently alter the physiological trajectory of brain development, we set out to investigate the long-term consequences on dendritic morphology of subicular pyramidal neurons and expression on genes regulating the complex neural processes such as neuronal connectivity, learning, and memory. Using a well-established model of anesthetic neurotoxicity in rats and mice neonatally exposed to sevoflurane, a volatile general anesthetic commonly used in pediatric anesthesia, we report that a single 6 h of continuous anesthesia administered at postnatal day (PND) 7 resulted in lasting dysregulation in subicular mRNA levels of cAMP responsive element modulator (Crem), cAMP responsive element-binding protein 1 (Creb1), and Protein phosphatase 3 catalytic subunit alpha, a subunit of calcineurin (Ppp3ca) (calcineurin) when examined during juvenile period at PND28. Given the critical role of these genes in synaptic development and neuronal plasticity, we deployed a set of histological measurements to investigate the implications of anesthesia-induced dysregulation of gene expression on morphology and complexity of surviving subicular pyramidal neurons. Our results indicate that neonatal exposure to sevoflurane induced lasting rearrangement of subicular dendrites, resulting in higher orders of complexity and increased branching with no significant effects on the soma of pyramidal neurons. Correspondingly, changes in dendritic complexity were paralleled by the increased spine density on apical dendrites, further highlighting the scope of anesthesia-induced dysregulation of synaptic development. We conclude that neonatal sevoflurane induced persistent genetic and morphological dysregulation in juvenile rodents, which could indicate heightened susceptibility toward cognitive and behavioral disorders we are beginning to recognize as sequelae of early-in-life anesthesia.
Collapse
Affiliation(s)
- Nemanja Useinovic
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Michelle Near
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Omar Hoseá Cabrera
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Annalisa Boscolo
- Institute of Anesthesia and Intensive Care, Padua University Hospital, Padua 35128. Italy
- Department of Medicine (DIMED), University of Padua, Padua 35128, Italy
| | - Andjelko Milosevic
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Rachel Harvey
- Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
| | - Adre Newson
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Shelby Chastain-Potts
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nidia Quillinan
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Neuronal Injury and Plasticity Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Vesna Jevtovic-Todorovic
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
4
|
Useinovic N, Jevtovic-Todorovic V. Controversies in Anesthesia-Induced Developmental Neurotoxicity. Best Pract Res Clin Anaesthesiol 2023. [DOI: 10.1016/j.bpa.2023.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
5
|
Cabrera OH, Useinovic N, Maksimovic S, Near M, Quillinan N, Todorovic SM, Jevtovic-Todorovic V. Neonatal ketamine exposure impairs infrapyramidal bundle pruning and causes lasting increase in excitatory synaptic transmission in hippocampal CA3 neurons. Neurobiol Dis 2022; 175:105923. [PMID: 36371060 PMCID: PMC9831613 DOI: 10.1016/j.nbd.2022.105923] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 10/27/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
Preclinical models demonstrate that nearly all anesthetics cause widespread neuroapoptosis in the developing brains of infant rodents and non-human primates. Anesthesia-induced developmental apoptosis is succeeded by prolonged neuropathology in the surviving neurons and lasting cognitive impairments, suggesting that anesthetics interfere with the normal developmental trajectory of the brain. However, little is known about effects of anesthetics on stereotyped axonal pruning, an important developmental algorithm that sculpts neural circuits for proper function. Here, we proposed that neonatal ketamine exposure may interfere with stereotyped axonal pruning of the infrapyramidal bundle (IPB) of the hippocampal mossy fiber system and that impaired pruning may be associated with alterations in the synaptic transmission of CA3 neurons. To test this hypothesis, we injected postnatal day 7 (PND7) mouse pups with ketamine or vehicle over 6 h and then studied them at different developmental stages corresponding to IPB pruning (PND20-40). Immunohistochemistry with synaptoporin (a marker of mossy fibers) revealed that in juvenile mice treated with ketamine at PND7, but not in vehicle-treated controls, positive IPB fibers extended farther into the stratum pyramidale of CA3 region. Furthermore, immunofluorescent double labeling for synaptoporin and PSD-95 strongly suggested that the unpruned IPB caused by neonatal ketamine exposure makes functional synapses. Importantly, patch-clamp electrophysiology for miniature excitatory postsynaptic currents (mEPSCs) in acute brain slices ex vivo revealed increased frequency and amplitudes of mEPSCs in hippocampal CA3 neurons in ketamine-treated groups when compared to vehicle controls. We conclude that neonatal ketamine exposure interferes with normal neural circuit development and that this interference leads to lasting increase in excitatory synaptic transmission in hippocampus.
Collapse
Affiliation(s)
- Omar Hoseá Cabrera
- University of Colorado School of Medicine at Anschutz Medical Campus, Department of Anesthesiology, Aurora, CO, USA
| | - Nemanja Useinovic
- University of Colorado School of Medicine at Anschutz Medical Campus, Department of Anesthesiology, Aurora, CO, USA
| | - Stefan Maksimovic
- University of Colorado School of Medicine at Anschutz Medical Campus, Department of Anesthesiology, Aurora, CO, USA
| | - Michelle Near
- University of Colorado School of Medicine at Anschutz Medical Campus, Department of Anesthesiology, Aurora, CO, USA
| | - Nidia Quillinan
- University of Colorado School of Medicine at Anschutz Medical Campus, Department of Anesthesiology, Aurora, CO, USA,University of Colorado School of Medicine at Anschutz Medical Campus, Neuroscience Graduate Program, Aurora, CO, USA
| | - Slobodan M. Todorovic
- University of Colorado School of Medicine at Anschutz Medical Campus, Department of Anesthesiology, Aurora, CO, USA,University of Colorado School of Medicine at Anschutz Medical Campus, Neuroscience Graduate Program, Aurora, CO, USA
| | - Vesna Jevtovic-Todorovic
- University of Colorado School of Medicine at Anschutz Medical Campus, Department of Anesthesiology, Aurora, CO, USA,University of Colorado School of Medicine at Anschutz Medical Campus, Department of Pharmacology, Aurora, CO, USA,Corresponding author. (V. Jevtovic-Todorovic)
| |
Collapse
|
6
|
Cheng Y, Liu S, Zhang L, Jiang H. Identification of Prefrontal Cortex and Amygdala Expressed Genes Associated With Sevoflurane Anesthesia on Non-human Primate. Front Integr Neurosci 2022; 16:857349. [PMID: 35845920 PMCID: PMC9286018 DOI: 10.3389/fnint.2022.857349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 05/19/2022] [Indexed: 11/22/2022] Open
Abstract
Clinical trials and animal studies have indicated that long-term use or multiple administrations of anesthesia may lead to fine motor impairment in the developing brain. Most studies on anesthesia-induced neurotoxicity have focused on the hippocampus and prefrontal cortex (PFC); however, the role of other vital encephalic regions, such as the amygdala, is still unclear. Herein, we focused on sevoflurane, the most commonly used volatile anesthetic in infants, and performed a transcriptional analysis of the PFC and amygdala of macaques after multiple exposures to the anesthetic by RNA sequencing. The overall, overlapping, and encephalic region-specific transcriptional patterns were separately analyzed to reveal their functions and differentially expressed gene sets that were influenced by sevoflurane. Specifically, functional, protein–protein interaction, neighbor gene network, and gene set enrichment analyses were performed. Further, we built the basic molecular feature of the amygdala by comparing it to the PFC. In comparison with the amygdala’s changing pattern following sevoflurane exposure, functional annotations of the PFC were more enriched in glial cell-related biological functions than in neuron and synapsis development. Taken together, transcriptional studies and bioinformatics analyses allow for an improved understanding of the primate PFC and amygdala.
Collapse
|
7
|
Turner AD, Sullivan T, Drury K, Hall TA, Williams CN, Guilliams KP, Murphy S, Iqbal O’Meara AM. Cognitive Dysfunction After Analgesia and Sedation: Out of the Operating Room and Into the Pediatric Intensive Care Unit. Front Behav Neurosci 2021; 15:713668. [PMID: 34483858 PMCID: PMC8415404 DOI: 10.3389/fnbeh.2021.713668] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/26/2021] [Indexed: 11/13/2022] Open
Abstract
In the midst of concerns for potential neurodevelopmental effects after surgical anesthesia, there is a growing awareness that children who require sedation during critical illness are susceptible to neurologic dysfunctions collectively termed pediatric post-intensive care syndrome, or PICS-p. In contrast to healthy children undergoing elective surgery, critically ill children are subject to inordinate neurologic stress or injury and need to be considered separately. Despite recognition of PICS-p, inconsistency in techniques and timing of post-discharge assessments continues to be a significant barrier to understanding the specific role of sedation in later cognitive dysfunction. Nonetheless, available pediatric studies that account for analgesia and sedation consistently identify sedative and opioid analgesic exposures as risk factors for both in-hospital delirium and post-discharge neurologic sequelae. Clinical observations are supported by animal models showing neuroinflammation, increased neuronal death, dysmyelination, and altered synaptic plasticity and neurotransmission. Additionally, intensive care sedation also contributes to sleep disruption, an important and overlooked variable during acute illness and post-discharge recovery. Because analgesia and sedation are potentially modifiable, understanding the underlying mechanisms could transform sedation strategies to improve outcomes. To move the needle on this, prospective clinical studies would benefit from cohesion with regard to datasets and core outcome assessments, including sleep quality. Analyses should also account for the wide range of diagnoses, heterogeneity of this population, and the dynamic nature of neurodevelopment in age cohorts. Much of the related preclinical evidence has been studied in comparatively brief anesthetic exposures in healthy animals during infancy and is not generalizable to critically ill children. Thus, complementary animal models that more accurately "reverse translate" critical illness paradigms and the effect of analgesia and sedation on neuropathology and functional outcomes are needed. This review explores the interactive role of sedatives and the neurologic vulnerability of critically ill children as it pertains to survivorship and functional outcomes, which is the next frontier in pediatric intensive care.
Collapse
Affiliation(s)
- Ashley D. Turner
- Division of Pediatric Critical Care, Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, United States
| | - Travis Sullivan
- Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Kurt Drury
- Department of Pediatrics, Division of Pediatric Critical Care, Doernbecher Children’s Hospital, Oregon Health & Science University, Portland, OR, United States
| | - Trevor A. Hall
- Department of Pediatrics, Division of Pediatric Psychology, Pediatric Critical Care and Neurotrauma Recovery Program, Doernbecher Children’s Hospital, Oregon Health & Science University, Portland, OR, United States
| | - Cydni N. Williams
- Department of Pediatrics, Division of Pediatric Critical Care, Doernbecher Children’s Hospital, Oregon Health & Science University, Portland, OR, United States
| | - Kristin P. Guilliams
- Division of Pediatric Critical Care, Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, United States
- Division of Pediatric Neurology, Department of Neurology, Washington University in St. Louis, St. Louis, MO, United States
- Division of Neuroradiology, Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, United States
| | - Sarah Murphy
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - A. M. Iqbal O’Meara
- Department of Pediatrics, Child Health Research Institute, Children’s Hospital of Richmond at Virginia Commonwealth University School of Medicine, Richmond, VA, United States
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
8
|
Cabrera OH, Useinovic N, Jevtovic-Todorovic V. Neonatal Anesthesia and dysregulation of the Epigenome. Biol Reprod 2021; 105:720-734. [PMID: 34258621 DOI: 10.1093/biolre/ioab136] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/30/2021] [Accepted: 07/09/2021] [Indexed: 11/14/2022] Open
Abstract
Each year, millions of infants and children are anesthetized for medical and surgical procedures. Yet, a substantial body of preclinical evidence suggests that anesthetics are neurotoxins that cause rapid and widespread apoptotic cell death in the brains of infant rodents and non-human primates. These animals have persistent impairments in cognition and behavior many weeks or months after anesthesia exposure, leading us to hypothesize that anesthetics do more than simply kill brain cells. Indeed, anesthetics cause chronic neuropathology in neurons that survive the insult, which then interferes with major aspects of brain development, synaptic plasticity, and neuronal function. Understanding the phenomenon of anesthesia-induced developmental neurotoxicity is of critical public health importance because clinical studies now report that anesthesia in human infancy is associated with cognitive and behavioral deficits. In our search for mechanistic explanations for why a young and pliable brain cannot fully recover from a relatively brief period of anesthesia, we have accumulated evidence that neonatal anesthesia can dysregulate epigenetic tags that influence gene transcription such as histone acetylation and DNA methylation. In this review, we briefly summarize the phenomenon of anesthesia-induced developmental neurotoxicity. We then discuss chronic neuropathology caused by neonatal anesthesia, including disturbances in cognition, socio-affective behavior, neuronal morphology, and synaptic plasticity. Finally, we present evidence of anesthesia-induced genetic and epigenetic dysregulation within the developing brain that may be transmitted intergenerationally to anesthesia-naïve offspring.
Collapse
Affiliation(s)
- Omar Hoseá Cabrera
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States of America
| | - Nemanja Useinovic
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States of America
| | - Vesna Jevtovic-Todorovic
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States of America
| |
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW Perinatal depression is a common and debilitating complication of pregnancy and childbirth. Recent studies have elucidated relationships between acute birth events on depression risk, and novel treatments for postpartum depression have been discovered and approved. This article reviews current understandings about birth events on depression, new screening standards, and novel treatments for postpartum depression. RECENT FINDINGS Pain, analgesia, and depression are complex traits that are inter-related during and after pregnancy. Certain individuals may benefit more than others from addressing pain and suffering around childbirth. Exposures to general anesthesia or postdural puncture headache are associated with postpartum depression symptoms, although a causal relationship is unlikely. Brexanolone, ketamine and its related compounds, and nonpharmacologic options offer new or alternative therapies for depression, although safety information for some of these treatments in pregnancy and lactation are needed. Maternal health bundles call for close attention to perinatal mental health screening with validated instruments, and for timely treatment referrals in the 'fourth trimester'. SUMMARY Clinical monitoring and timely treatment of depression in the perinatal and postpartum periods is critical for maternal postpartum health and recovery. Perinatal specialists and researchers should continue to focus on tailored treatments specific to this special population.
Collapse
Affiliation(s)
- Grace Lim
- University of Pittsburgh Department of Anesthesiology & Perioperative Medicine
- University of Pittsburgh Department of Obstetrics & Gynecology, UPMC Magee-Women’s Hospital
| |
Collapse
|
10
|
Xie L, Hu Y, Yan D, McQuillan P, Liu Y, Zhu S, Zhu Z, Jiang Y, Hu Z. The relationship between exposure to general anesthetic agents and the risk of developing an impulse control disorder. Pharmacol Res 2021; 165:105440. [PMID: 33493656 DOI: 10.1016/j.phrs.2021.105440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/13/2021] [Accepted: 01/16/2021] [Indexed: 12/18/2022]
Abstract
Most studies examining the effect of extended exposure to general anesthetic agents (GAAs) have demonstrated that extended exposure induces both structural and functional changes in the central nervous system. These changes are frequently accompanied by neurobehavioral changes that include impulse control disorders that are generally characterized by deficits in behavioral inhibition and executive function. In this review, we will.
Collapse
Affiliation(s)
- Linghua Xie
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuhan Hu
- Department of Cell Biology, Yale University, New Haven, CT, USA
| | - Dandan Yan
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - P McQuillan
- Department of Anesthesiology, Penn State Hershey Medical Centre, Penn State College of Medicine, Hershey, PA, USA
| | - Yue Liu
- Department of Anesthesiology, The Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shengmei Zhu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhirui Zhu
- Department of Anesthesiology, The Children Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yilei Jiang
- Department of Anesthesiology, The Children Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiyong Hu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
11
|
Slupe AM, Villasana L, Wright KM. GABAergic neurons are susceptible to BAX-dependent apoptosis following isoflurane exposure in the neonatal period. PLoS One 2021; 16:e0238799. [PMID: 33434191 PMCID: PMC7802958 DOI: 10.1371/journal.pone.0238799] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/27/2020] [Indexed: 12/18/2022] Open
Abstract
Exposure to volatile anesthetics during the neonatal period results in acute neuron death. Prior work suggests that apoptosis is the dominant mechanism mediating neuron death. We show that Bax deficiency blocks neuronal death following exposure to isoflurane during the neonatal period. Blocking Bax-mediated neuron death attenuated the neuroinflammatory response of microglia following isoflurane exposure. We find that GABAergic interneurons are disproportionately overrepresented among dying neurons. Despite the increase in neuronal apoptosis induced by isoflurane exposure during the neonatal period, seizure susceptibility, spatial memory retention, and contextual fear memory were unaffected later in life. However, Bax deficiency alone led to mild deficiencies in spatial memory and contextual fear memory, suggesting that normal developmental apoptotic death is important for cognitive function. Collectively, these findings show that while GABAergic neurons in the neonatal brain undergo elevated Bax-dependent apoptotic cell death following exposure to isoflurane, this does not appear to have long-lasting consequences on overall neurological function later in life.
Collapse
Affiliation(s)
- Andrew M. Slupe
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Laura Villasana
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Kevin M. Wright
- Vollum Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- * E-mail:
| |
Collapse
|
12
|
Ketamine inhibits neuronal differentiation by regulating brain-derived neurotrophic factor (BDNF) signaling. Toxicol In Vitro 2021; 72:105091. [PMID: 33440188 DOI: 10.1016/j.tiv.2021.105091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 11/28/2020] [Accepted: 01/07/2021] [Indexed: 11/22/2022]
Abstract
Ketamine is widely used in pediatric anesthesia, perioperative sedation, and analgesia. Knowledge of anesthesia neurotoxicity in humans is currently limited by the difficulty of obtaining neurons and performing developmental toxicity studies in fetal and pediatric populations. However, mouse embryonic stem cells (mESCs) derived from embryos at the preimplantation stage demonstrate an unlimited ability to self-renew and generate different cell types and are a valuable tool for clinical research. Thus, in this study, a model was employed to investigate the mechanism by which ketamine (200 nM) influences the neuronal differentiation of mESCs. Mouse ESCs were treated with an anesthetic dose of ketamine, and neuronal differentiation was significantly inhibited on day 5. Downregulation of brain-derived neurotrophic factor (BDNF) by shRNA was found to have the same inhibitory effect. Furthermore, a rescue experiment indicated that BDNF overexpression markedly restored the neuronal differentiation inhibited by ketamine in the ketamine/BDNF group on day 5. Taken together, these data suggested that ketamine inhibited the neuronal differentiation of mESCs, possibly by interfering with BDNF. The results of the current study may provide novel ideas for preventing ketamine toxicity in the developing fetus.
Collapse
|
13
|
Lyu D, Tang N, Womack AW, He YJ, Lin Q. Neonatal ketamine exposure-induced hippocampal neuroapoptosis in the developing brain impairs adult spatial learning ability. Neural Regen Res 2020; 15:880-886. [PMID: 31719253 PMCID: PMC6990767 DOI: 10.4103/1673-5374.268929] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 02/25/2019] [Accepted: 06/10/2019] [Indexed: 12/19/2022] Open
Abstract
Ketamine exposure can lead to selective neuroapoptosis in the developing brain. p66ShcA, the cellular adapter protein expressed selectively in immature neurons, is a known pro-apoptotic molecule that triggers neuroapoptosis when activated. Sprague-Dawley rats at postnatal day 7 were subcutaneously injected in the neck with ketamine 20 mg/kg, six times at 2-hour intervals. At 0, 1, 3, and 6 hours after final injection, western blot assay was used to detect the expression of cleaved caspase-3, p66ShcA, and phosphorylated p66ShcA. We found that the expression of activated p66ShcA and caspase-3 increased after ketamine exposure and peaked at 3 hours. The same procedure was performed on a different group of rats. At the age of 4 weeks, spatial learning and memory abilities were tested with the Morris water maze. Latency to find the hidden platform for these rats was longer than it was for control rats, although the residence time in the target quadrant was similar. These findings indicate that ketamine exposure resulted in p66ShcA being activated in the course of an apoptotic cascade during the neonatal period. This may have contributed to the deficit in spatial learning and memory that persisted into adulthood. The experimental protocol was approved by the Institutional Animal Care and Use Committee at the University of Texas at Arlington, USA (approval No. A13.008) on January 22, 2013.
Collapse
Affiliation(s)
- Dan Lyu
- Department of Pain Management, Tianjin First Center Hospital, Tianjin, China; Department of Psychology, College of Science, University of Texas at Arlington, Arlington, TX, USA
| | - Ning Tang
- Department of Psychology, College of Science, University of Texas at Arlington, Arlington, TX, USA; Reproductive Medicine Center, the 960th Hospital of the PLA Joint Logistics Support Force, Jinan, Shandong Province, China
| | - Andrew W Womack
- Department of Psychology, College of Science, University of Texas at Arlington, Arlington, TX, USA
| | - Yong-Jin He
- Department of Pain Management, Tianjin First Center Hospital, Tianjin, China
| | - Qing Lin
- Department of Psychology, College of Science, University of Texas at Arlington, Arlington, TX, USA
| |
Collapse
|
14
|
Potential Neurodevelopmental Effects of Pediatric Intensive Care Sedation and Analgesia: Repetitive Benzodiazepine and Opioid Exposure Alters Expression of Glial and Synaptic Proteins in Juvenile Rats. Crit Care Explor 2020; 2:e0105. [PMID: 32426747 PMCID: PMC7188419 DOI: 10.1097/cce.0000000000000105] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Sedatives are suspected contributors to neurologic dysfunction in PICU patients, to whom they are administered during sensitive neurodevelopment. Relevant preclinical modeling has largely used comparatively brief anesthesia in infant age-approximate animals, with insufficient study of repetitive combined drug administration during childhood. We hypothesized that childhood neurodevelopment is selectively vulnerable to repeated treatment with benzodiazepine and opioid. We report a preclinical model of combined midazolam and morphine in early childhood age-approximate rats.
Collapse
|
15
|
Ding R, Tan Y, Du A, Wen G, Ren X, Yao H, Ren W, Liu H, Wang X, Yu H, Yao J, Li B, Zhang G, Lu Y, Wu X. Redistribution of Monocarboxylate 1 and 4 in Hippocampus and Spatial Memory Impairment Induced by Long-term Ketamine Administration. Front Behav Neurosci 2020; 14:60. [PMID: 32362817 PMCID: PMC7181955 DOI: 10.3389/fnbeh.2020.00060] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 03/30/2020] [Indexed: 12/15/2022] Open
Abstract
The monocarboxylate transporters (MCTs) MCT1, MCT2, and MCT4 are essential components of the astrocyte-neuron lactate shuttle (ANLS), which is a fundamental element of brain energetics. Decreased expression of MCTs can induce cognitive dysfunction of the brain. In the present study, we established a mouse model of long-term ketamine administration by subjecting mice to a 6-month course of a daily intraperitoneal injection of ketamine. These mice demonstrated learning and memory deficits and a significant decline in MCT1 and MCT4 proteins in the hippocampal membrane fraction, while cytoplasmic MCT1 and MCT4 protein levels were significantly increased. In contrast, the levels of global MCT2 protein were significantly increased. Analysis of mRNA levels found no changes in MCT1/4 transcripts, although the expression of MCT2 mRNA was significantly increased. We suggest that redistribution of hippocampal MCT1 and MCT4, but not MCT2 up-regulation, may be related to learning and memory deficits induced by long-term ketamine administration.
Collapse
Affiliation(s)
- Runtao Ding
- School of Forensic Medicine, China Medical University, Shenyang, China.,Department of Forensic and Medical Laboratory, Jining Medical University, Jining, China
| | - Yaqing Tan
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Ao Du
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Gehua Wen
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Xinghua Ren
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Hui Yao
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Weishu Ren
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Huairu Liu
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Xiaolong Wang
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Hao Yu
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Jun Yao
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Baoman Li
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Guohua Zhang
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Yan Lu
- Key Laboratory of Health Ministry in Congenital Malformation, The Affiliated Shengjing Hospital of China Medical University, Shenyang, China
| | - Xu Wu
- School of Forensic Medicine, China Medical University, Shenyang, China
| |
Collapse
|
16
|
Costa G, De Luca MA, Piras G, Marongiu J, Fattore L, Simola N. Neuronal and peripheral damages induced by synthetic psychoactive substances: an update of recent findings from human and animal studies. Neural Regen Res 2020; 15:802-816. [PMID: 31719240 PMCID: PMC6990793 DOI: 10.4103/1673-5374.268895] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Preclinical and clinical studies indicate that synthetic psychoactive substances, in addition to having abuse potential, may elicit toxic effects of varying severity at the peripheral and central levels. Nowadays, toxicity induced by synthetic psychoactive substances poses a serious harm for health, since recreational use of these substances is on the rise among young and adult people. The present review summarizes recent findings on the peripheral and central toxicity elicited by “old” and “new” synthetic psychoactive substances in humans and experimental animals, focusing on amphetamine derivatives, hallucinogen and dissociative drugs and synthetic cannabinoids.
Collapse
Affiliation(s)
- Giulia Costa
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Maria Antonietta De Luca
- Department of Biomedical Sciences; National Institute of Neuroscience (INN), University of Cagliari, Cagliari, Italy
| | - Gessica Piras
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Jacopo Marongiu
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Liana Fattore
- National Research Council of Italy, Institute of Neuroscience, Cagliari, Italy
| | - Nicola Simola
- Department of Biomedical Sciences; National Institute of Neuroscience (INN), University of Cagliari, Cagliari, Italy
| |
Collapse
|
17
|
General anesthetic exposure in adolescent rats causes persistent maladaptations in cognitive and affective behaviors and neuroplasticity. Neuropharmacology 2019; 150:153-163. [PMID: 30926450 DOI: 10.1016/j.neuropharm.2019.03.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 03/15/2019] [Accepted: 03/15/2019] [Indexed: 01/25/2023]
Abstract
Accumulating evidence indicates that exposure to general anesthetics during infancy and childhood can cause persistent cognitive impairment, alterations in synaptic plasticity, and, to a lesser extent, increased incidence of behavioral disorders. Unfortunately, the developmental parameters of susceptibility to general anesthetics are not well understood. Adolescence is a critical developmental period wherein multiple late developing brain regions may also be vulnerable to enduring general anesthetic effects. Given the breadth of the adolescent age span, this group potentially represents millions more individuals than those exposed during early childhood. In this study, isoflurane exposure within a well-characterized adolescent period in Sprague-Dawley rats elicited immediate and persistent anxiety- and impulsive-like responding, as well as delayed cognitive impairment into adulthood. These behavioral abnormalities were paralleled by atypical dendritic spine morphology in the prefrontal cortex (PFC) and hippocampus (HPC), suggesting delayed anatomical maturation, and shifts in inhibitory function that suggest hypermaturation of extrasynaptic GABAA receptor inhibition. Preventing this hypermaturation of extrasynaptic GABAA receptor-mediated function in the PFC selectively reversed enhanced impulsivity resulting from adolescent isoflurane exposure. Taken together, these data demonstrate that the developmental window for susceptibility to enduring untoward effects of general anesthetics may be much longer than previously appreciated, and those effects may include affective behaviors in addition to cognition.
Collapse
|
18
|
Limb Remote Ischemic Preconditioning Reduces Repeated Ketamine Exposure-Induced Adverse Effects in the Developing Brain of Rats. J Mol Neurosci 2019; 68:58-65. [DOI: 10.1007/s12031-019-01282-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 02/18/2019] [Indexed: 11/26/2022]
|
19
|
Maloney SE, Yuede CM, Creeley CE, Williams SL, Huffman JN, Taylor GT, Noguchi KN, Wozniak DF. Repeated neonatal isoflurane exposures in the mouse induce apoptotic degenerative changes in the brain and relatively mild long-term behavioral deficits. Sci Rep 2019; 9:2779. [PMID: 30808927 PMCID: PMC6391407 DOI: 10.1038/s41598-019-39174-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 01/18/2019] [Indexed: 11/22/2022] Open
Abstract
Epidemiological studies suggest exposures to anesthetic agents and/or sedative drugs (AASDs) in children under three years old, or pregnant women during the third trimester, may adversely affect brain development. Evidence suggests lengthy or repeated AASD exposures are associated with increased risk of neurobehavioral deficits. Animal models have been valuable in determining the type of acute damage in the developing brain induced by AASD exposures, as well as in elucidating long-term functional consequences. Few studies examining very early exposure to AASDs suggest this may be a critical period for inducing long-term functional consequences, but the impact of repeated exposures at these ages has not yet been assessed. To address this, we exposed mouse pups to a prototypical general anesthetic, isoflurane (ISO, 1.5% for 3 hr), at three early postnatal ages (P3, P5 and P7). We quantified the acute neuroapoptotic response to a single versus repeated exposure, and found age- and brain region-specific effects. We also found that repeated early exposures to ISO induced subtle, sex-specific disruptions to activity levels, motor coordination, anxiety-related behavior and social preference. Our findings provide evidence that repeated ISO exposures may induce behavioral disturbances that are subtle in nature following early repeated exposures to a single AASD.
Collapse
Affiliation(s)
- Susan E Maloney
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Psychology, University of Missouri - St. Louis, St. Louis, MO, 63121, USA
- Intellectual and Developmental Disabilities Research Center, Washington University, St. Louis, MO, USA
| | - Carla M Yuede
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Catherine E Creeley
- Department of Psychology, State University of New York at Fredonia, Fredonia, NY, 14063, USA
| | - Sasha L Williams
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jacob N Huffman
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - George T Taylor
- Department of Psychology, University of Missouri - St. Louis, St. Louis, MO, 63121, USA
| | - Kevin N Noguchi
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Intellectual and Developmental Disabilities Research Center, Washington University, St. Louis, MO, USA
| | - David F Wozniak
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA.
- Intellectual and Developmental Disabilities Research Center, Washington University, St. Louis, MO, USA.
| |
Collapse
|
20
|
Pearn ML, Schilling JM, Jian M, Egawa J, Wu C, Mandyam CD, Fannon-Pavlich MJ, Nguyen U, Bertoglio J, Kodama M, Mahata SK, DerMardirossian C, Lemkuil BP, Han R, Mobley WC, Patel HH, Patel PM, Head BP. Inhibition of RhoA reduces propofol-mediated growth cone collapse, axonal transport impairment, loss of synaptic connectivity, and behavioural deficits. Br J Anaesth 2018; 120:745-760. [PMID: 29576115 PMCID: PMC6200100 DOI: 10.1016/j.bja.2017.12.033] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/28/2017] [Accepted: 12/26/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Exposure of the developing brain to propofol results in cognitive deficits. Recent data suggest that inhibition of neuronal apoptosis does not prevent cognitive defects, suggesting mechanisms other than neuronal apoptosis play a role in anaesthetic neurotoxicity. Proper neuronal growth during development is dependent upon growth cone morphology and axonal transport. Propofol modulates actin dynamics in developing neurones, causes RhoA-dependent depolymerisation of actin, and reduces dendritic spines and synapses. We hypothesised that RhoA inhibition prevents synaptic loss and subsequent cognitive deficits. The present study tested whether RhoA inhibition with the botulinum toxin C3 (TAT-C3) prevents propofol-induced synapse and neurite loss, and preserves cognitive function. METHODS RhoA activation, growth cone morphology, and axonal transport were measured in neonatal rat neurones (5-7 days in vitro) exposed to propofol. Synapse counts (electron microscopy), dendritic arborisation (Golgi-Cox), and network connectivity were measured in mice (age 28 days) previously exposed to propofol at postnatal day 5-7. Memory was assessed in adult mice (age 3 months) previously exposed to propofol at postnatal day 5-7. RESULTS Propofol increased RhoA activation, collapsed growth cones, and impaired retrograde axonal transport of quantum dot-labelled brain-derived neurotrophic factor, all of which were prevented with TAT-C3. Adult mice previously treated with propofol had decreased numbers of total hippocampal synapses and presynaptic vesicles, reduced hippocampal dendritic arborisation, and infrapyramidal mossy fibres. These mice also exhibited decreased hippocampal-dependent contextual fear memory recall. All anatomical and behavioural changes were prevented with TAT-C3 pre-treatment. CONCLUSION Inhibition of RhoA prevents propofol-mediated hippocampal neurotoxicity and associated cognitive deficits.
Collapse
Affiliation(s)
- M L Pearn
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - J M Schilling
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - M Jian
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA; Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - J Egawa
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - C Wu
- Department of Neurosciences, UCSD, San Diego, CA, USA
| | - C D Mandyam
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - M J Fannon-Pavlich
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - U Nguyen
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - J Bertoglio
- INSERM U749, Institut Gustave Roussy, Universite Paris-sud, Paris, France
| | - M Kodama
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA; Metabolic Physiology and Ultrastructural Biology Laboratory, UCSD, San Diego CA, USA; Department of Anesthesiology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - S K Mahata
- Metabolic Physiology and Ultrastructural Biology Laboratory, UCSD, San Diego CA, USA
| | - C DerMardirossian
- Department of Immunology and Microbial Sciences, TSRI, La Jolla, CA, USA; Department of Cell and Molecular Biology, TSRI, La Jolla, CA, USA
| | - B P Lemkuil
- Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - R Han
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - W C Mobley
- Department of Neurosciences, UCSD, San Diego, CA, USA
| | - H H Patel
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - P M Patel
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - B P Head
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA.
| |
Collapse
|