1
|
Liu P, Li Q, Tang YF, Cui CY, Liu Q, Zhang Y, Tang B, Lai QC. Multiple algorithms highlight key brain genes driven by multiple anesthetics. Comput Biol Med 2024; 179:108805. [PMID: 38991319 DOI: 10.1016/j.compbiomed.2024.108805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/05/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024]
Abstract
Anesthesia serves as a pivotal tool in modern medicine, creating a transient state of sensory deprivation to ensure a pain-free surgical or medical intervention. While proficient in alleviating pain, anesthesia significantly modulates brain dynamics, metabolic processes, and neural signaling, thereby impairing typical cognitive functions. Furthermore, anesthesia can induce notable impacts such as memory impairment, decreased cognitive function, and diminished intelligence, emphasizing the imperative need to explore the concealed repercussions of anesthesia on individuals. In this investigation, we aggregated gene expression profiles (GSE64617, GSE141242, GSE161322, GSE175894, and GSE178995) from public repositories following second-generation sequencing analysis of various anesthetics. Through scrutinizing post-anesthesia brain tissue gene expression utilizing Gene Set Enrichment Analysis (GSEA), Robust Rank Aggregation (RRA), and Weighted Gene Co-expression Network Analysis (WGCNA), this research aims to pinpoint pivotal genes, pathways, and regulatory networks linked to anesthesia. This undertaking not only enhances comprehension of the physiological changes brought about by anesthesia but also lays the groundwork for future investigations, cultivating new insights and innovative perspectives in medical practice.
Collapse
Affiliation(s)
- Ping Liu
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Qun Li
- Department of Pain, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yi-Fan Tang
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Chun-Yan Cui
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China; Department of Pain, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Qing Liu
- Department of Pain, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China; Department of Anesthesiology, Hejiang Hospital of Traditional Chinese Medicine, Southwest Medical University, China
| | - Ying Zhang
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China; Central Nervous System Drug Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou, 646000, Sichuan, China; Department of Anesthesiology, Hejiang Hospital of Traditional Chinese Medicine, Southwest Medical University, China.
| | - Bo Tang
- Department of Pathology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Qian-Cheng Lai
- Department of Cardiac Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
2
|
Santandrea E, Aliakbari F, Truscott E, McCaig L, Donison NS, Graham D, Strong MJ, Volkening K. A technique for repeated blood and cerebrospinal fluid sampling from individual rats over time without the need for repeated anesthesia. Sci Rep 2024; 14:5171. [PMID: 38431711 PMCID: PMC10908789 DOI: 10.1038/s41598-024-55666-6] [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: 09/06/2023] [Accepted: 02/26/2024] [Indexed: 03/05/2024] Open
Abstract
Ethical animal use follows the 3R's: Replacement, Reduction and Refinement. Here, we present the use of simultaneous jugular vein and cisterna magna catheterization via a port system in rats for repeated fluid sampling for 14 consecutive days without loss of catheter patency. This technique allows repeated intra-animal sampling without anesthesia and, if used with pooling samples from a cohort of animals, replaces the need for terminal collections for sufficient sample volumes.
Collapse
Affiliation(s)
- Erin Santandrea
- Molecular Medicine, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Farhang Aliakbari
- Molecular Medicine, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Emily Truscott
- Animal Care and Veterinary Services, University of Western Ontario, London, Canada
| | - Lynda McCaig
- Molecular Medicine, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Neil S Donison
- Molecular Medicine, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Danielle Graham
- Molecular Medicine, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Michael J Strong
- Molecular Medicine, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada.
- Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.
| | - Kathryn Volkening
- Molecular Medicine, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
- Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
3
|
Alharbi KS, Almalki WH, Alzarea SI, Kazmi I, Al-Abbasi FA, Afzal O, Altamimi ASA, Albratty M, Najmi A, Gupta G. Anaesthesia-induced Changes in Genomic Expression Leading to Neurodegeneration. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:411-419. [PMID: 37157197 DOI: 10.2174/1871527322666230508123558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 05/10/2023]
Abstract
General anaesthetics (GA) have been in continuous clinical use for more than 170 years, with millions of young and elderly populations exposed to GA to relieve perioperative discomfort and carry out invasive examinations. Preclinical studies have shown that neonatal rodents with acute and chronic exposure to GA suffer from memory and learning deficits, likely due to an imbalance between excitatory and inhibitory neurotransmitters, which has been linked to neurodevelopmental disorders. However, the mechanisms behind anaesthesia-induced alterations in late postnatal mice have yet to be established. In this narrative review, we present the current state of knowledge on early life anaesthesia exposure-mediated alterations of genetic expression, focusing on insights gathered on propofol, ketamine, and isoflurane, as well as the relationship between network effects and subsequent biochemical changes that lead to long-term neurocognitive abnormalities. Our review provides strong evidence and a clear picture of anaesthetic agents' pathological events and associated transcriptional changes, which will provide new insights for researchers to elucidate the core ideas and gain an in-depth understanding of molecular and genetic mechanisms. These findings are also helpful in generating more evidence for understanding the exacerbated neuropathology, impaired cognition, and LTP due to acute and chronic exposure to anaesthetics, which will be beneficial for the prevention and treatment of many diseases, such as Alzheimer's disease. Given the many procedures in medical practice that require continuous or multiple exposures to anaesthetics, our review will provide great insight into the possible adverse impact of these substances on the human brain and cognition.
Collapse
Affiliation(s)
- Khalid Saad Alharbi
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fahad A Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, 11942, Saudi Arabia
| | | | - Mohammed Albratty
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box. 114, Jazan 45142, Saudi Arabia
| | - Asim Najmi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box. 114, Jazan 45142, Saudi Arabia
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, 302017, Jaipur, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| |
Collapse
|
4
|
Dell’Orco M, Weisend JE, Perrone-Bizzozero NI, Carlson AP, Morton RA, Linsenbardt DN, Shuttleworth CW. Repetitive spreading depolarization induces gene expression changes related to synaptic plasticity and neuroprotective pathways. Front Cell Neurosci 2023; 17:1292661. [PMID: 38162001 PMCID: PMC10757627 DOI: 10.3389/fncel.2023.1292661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/17/2023] [Indexed: 01/03/2024] Open
Abstract
Spreading depolarization (SD) is a slowly propagating wave of profound depolarization that sweeps through cortical tissue. While much emphasis has been placed on the damaging consequences of SD, there is uncertainty surrounding the potential activation of beneficial pathways such as cell survival and plasticity. The present study used unbiased assessments of gene expression to evaluate that compensatory and repair mechanisms could be recruited following SD, regardless of the induction method, which prior to this work had not been assessed. We also tested assumptions of appropriate controls and the spatial extent of expression changes that are important for in vivo SD models. SD clusters were induced with either KCl focal application or optogenetic stimulation in healthy mice. Cortical RNA was extracted and sequenced to identify differentially expressed genes (DEGs). SDs using both induction methods significantly upregulated 16 genes (vs. sham animals) that included the cell proliferation-related genes FOS, JUN, and DUSP6, the plasticity-related genes ARC and HOMER1, and the inflammation-related genes PTGS2, EGR2, and NR4A1. The contralateral hemisphere is commonly used as control tissue for DEG studies, but its activity could be modified by near-global disruption of activity in the adjacent brain. We found 21 upregulated genes when comparing SD-involved cortex vs. tissue from the contralateral hemisphere of the same animals. Interestingly, there was almost complete overlap (21/16) with the DEGs identified using sham controls. Neuronal activity also differs in SD initiation zones, where sustained global depolarization is required to initiate propagating events. We found that gene expression varied as a function of the distance from the SD initiation site, with greater expression differences observed in regions further away. Functional and pathway enrichment analyses identified axonogenesis, branching, neuritogenesis, and dendritic growth as significantly enriched in overlapping DEGs. Increased expression of SD-induced genes was also associated with predicted inhibition of pathways associated with cell death, and apoptosis. These results identify novel biological pathways that could be involved in plasticity and/or circuit modification in brain tissue impacted by SD. These results also identify novel functional targets that could be tested to determine potential roles in the recovery and survival of peri-infarct tissues.
Collapse
Affiliation(s)
- Michela Dell’Orco
- Department of Neurosciences, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Jordan E. Weisend
- Department of Neurosciences, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Nora I. Perrone-Bizzozero
- Department of Neurosciences, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Andrew P. Carlson
- Department of Neurosurgery, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Russell A. Morton
- Department of Neurosciences, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - David N. Linsenbardt
- Department of Neurosciences, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - C. William Shuttleworth
- Department of Neurosciences, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| |
Collapse
|
5
|
Jiwaji Z, Márkus NM, McQueen J, Emelianova K, He X, Dando O, Chandran S, Hardingham GE. General anesthesia alters CNS and astrocyte expression of activity-dependent and activity-independent genes. FRONTIERS IN NETWORK PHYSIOLOGY 2023; 3:1216366. [PMID: 37670849 PMCID: PMC10476527 DOI: 10.3389/fnetp.2023.1216366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/21/2023] [Indexed: 09/07/2023]
Abstract
General anesthesia represents a common clinical intervention and yet can result in long-term adverse CNS effects particularly in the elderly or dementia patients. Suppression of cortical activity is a key feature of the anesthetic-induced unconscious state, with activity being a well-described regulator of pathways important for brain health. However, the extent to which the effects of anesthesia go beyond simple suppression of neuronal activity is incompletely understood. We found that general anesthesia lowered cortical expression of genes induced by physiological activity in vivo, and recapitulated additional patterns of gene regulation induced by total blockade of firing activity in vitro, including repression of neuroprotective genes and induction of pro-apoptotic genes. However, the influence of anesthesia extended beyond that which could be accounted for by activity modulation, including the induction of non activity-regulated genes associated with inflammation and cell death. We next focused on astrocytes, important integrators of both neuronal activity and inflammatory signaling. General anesthesia triggered gene expression changes consistent with astrocytes being in a low-activity environment, but additionally caused induction of a reactive profile, with transcriptional changes enriched in those triggered by stroke, neuroinflammation, and Aß/tau pathology. Thus, while the effects of general anesthesia on cortical gene expression are consistent with the strong repression of brain activity, further deleterious effects are apparent including a reactive astrocyte profile.
Collapse
Affiliation(s)
- Zoeb Jiwaji
- UK Dementia Research Institute, Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Department of Anaesthesia, Critical Care and Pain Medicine, Usher Institute, Edinburgh Royal Infirmary, Edinburgh, United Kingdom
| | - Nóra M. Márkus
- UK Dementia Research Institute, Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Jamie McQueen
- UK Dementia Research Institute, Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Katie Emelianova
- UK Dementia Research Institute, Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Xin He
- UK Dementia Research Institute, Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Owen Dando
- UK Dementia Research Institute, Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Siddharthan Chandran
- UK Dementia Research Institute, Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Giles E. Hardingham
- UK Dementia Research Institute, Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
6
|
Jimenez-Tellez N, Pehar M, Visser F, Casas-Ortiz A, Rice T, Syed NI. Sevoflurane Exposure in Neonates Perturbs the Expression Patterns of Specific Genes That May Underly the Observed Learning and Memory Deficits. Int J Mol Sci 2023; 24:ijms24108696. [PMID: 37240038 DOI: 10.3390/ijms24108696] [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: 04/01/2023] [Revised: 04/20/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Exposure to commonly used anesthetics leads to neurotoxic effects in animal models-ranging from cell death to learning and memory deficits. These neurotoxic effects invoke a variety of molecular pathways, exerting either immediate or long-term effects at the cellular and behavioural levels. However, little is known about the gene expression changes following early neonatal exposure to these anesthetic agents. We report here on the effects of sevoflurane, a commonly used inhalational anesthetic, on learning and memory and identify a key set of genes that may likely be involved in the observed behavioural deficits. Specifically, we demonstrate that sevoflurane exposure in postnatal day 7 (P7) rat pups results in subtle, but distinct, memory deficits in the adult animals that have not been reported previously. Interestingly, when given intraperitoneally, pre-treatment with dexmedetomidine (DEX) could only prevent sevoflurane-induced anxiety in open field testing. To identify genes that may have been altered in the neonatal rats after sevoflurane and DEX exposure, specifically those impacting cellular viability, learning, and memory, we conducted an extensive Nanostring study examining over 770 genes. We found differential changes in the gene expression levels after exposure to both agents. A number of the perturbed genes found in this study have previously been implicated in synaptic transmission, plasticity, neurogenesis, apoptosis, myelination, and learning and memory. Our data thus demonstrate that subtle, albeit long-term, changes observed in an adult animal's learning and memory after neonatal anesthetic exposure may likely involve perturbation of specific gene expression patterns.
Collapse
Affiliation(s)
- Nerea Jimenez-Tellez
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Marcus Pehar
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Frank Visser
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Alberto Casas-Ortiz
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB T2N 4N1, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Tiffany Rice
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Anesthesiology, Perioperative and Pain Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Naweed I Syed
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
7
|
Coliță CI, Olaru DG, Coliță D, Hermann DM, Coliță E, Glavan D, Popa-Wagner A. Induced Coma, Death, and Organ Transplantation: A Physiologic, Genetic, and Theological Perspective. Int J Mol Sci 2023; 24:ijms24065744. [PMID: 36982814 PMCID: PMC10059721 DOI: 10.3390/ijms24065744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
In the clinic, the death certificate is issued if brain electrical activity is no longer detectable. However, recent research has shown that in model organisms and humans, gene activity continues for at least 96 h postmortem. The discovery that many genes are still working up to 48 h after death questions our definition of death and has implications for organ transplants and forensics. If genes can be active up to 48 h after death, is the person technically still alive at that point? We discovered a very interesting parallel between genes that were upregulated in the brain after death and genes upregulated in the brains that were subjected to medically-induced coma, including transcripts involved in neurotransmission, proteasomal degradation, apoptosis, inflammation, and most interestingly, cancer. Since these genes are involved in cellular proliferation, their activation after death could represent the cellular reaction to escape mortality and raises the question of organ viability and genetics used for transplantation after death. One factor limiting the organ availability for transplantation is religious belief. However, more recently, organ donation for the benefit of humans in need has been seen as “posthumous giving of organs and tissues can be a manifestation of love spreading also to the other side of death”.
Collapse
Affiliation(s)
- Cezar-Ivan Coliță
- Doctoral School, University of Medicine and Pharmacy Carol Davila, 020276 Bucharest, Romania; (C.-I.C.)
| | - Denissa-Greta Olaru
- Department of Psychiatry, University for Medicine and Pharmacy Craiova, 200349 Craiova, Romania;
| | - Daniela Coliță
- Doctoral School, University of Medicine and Pharmacy Carol Davila, 020276 Bucharest, Romania; (C.-I.C.)
| | - Dirk M. Hermann
- Chair of Vascular Neurology, Dementia and Ageing, Department of Neurology, University Hospital Essen, 45147 Essen, Germany
| | - Eugen Coliță
- Doctoral School, University of Medicine and Pharmacy Carol Davila, 020276 Bucharest, Romania; (C.-I.C.)
| | - Daniela Glavan
- Department of Psychiatry, University for Medicine and Pharmacy Craiova, 200349 Craiova, Romania;
- Correspondence: (D.G.); (A.P.-W.)
| | - Aurel Popa-Wagner
- Department of Psychiatry, University for Medicine and Pharmacy Craiova, 200349 Craiova, Romania;
- Chair of Vascular Neurology, Dementia and Ageing, Department of Neurology, University Hospital Essen, 45147 Essen, Germany
- Correspondence: (D.G.); (A.P.-W.)
| |
Collapse
|
8
|
Davis JA, Grau JW. Protecting the injured central nervous system: Do anesthesia or hypothermia ameliorate secondary injury? Exp Neurol 2023; 363:114349. [PMID: 36775099 DOI: 10.1016/j.expneurol.2023.114349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/13/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023]
Abstract
Traumatic injury to the central nervous system (CNS) and stroke initiate a cascade of processes that expand the area of tissue loss. The current review considers recent studies demonstrating that the induction of an anesthetic state or cooling the affected tissue (hypothermia) soon after injury can have a therapeutic effect. We first provide an overview of the neurobiological processes that fuel tissue loss after traumatic brain injury (TBI), spinal cord injury (SCI) and stroke. We then examine the rehabilitative effectiveness of therapeutic anesthesia across a variety of drug categories through a systematic review of papers in the PubMed database. We also review the therapeutic benefits hypothermia, another treatment that quells neural activity. We conclude by considering factors related to the safety, efficacy and timing of treatment, as well as the mechanisms of action. Clinical implications are also discussed.
Collapse
Affiliation(s)
- Jacob A Davis
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA.
| | - James W Grau
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
9
|
Finamor F, Scarabelot VL, Medeiros LF, Stein DJ, da Silva MD, Callai E, Caumo W, de Souza A, Torres ILS. Involvement of GABAergic, glutamatergic, opioidergic, and brain-derived neurotrophic factor systems in the trigeminal neuropathic pain process. Neurosci Lett 2023; 793:136970. [PMID: 36402255 DOI: 10.1016/j.neulet.2022.136970] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
Abstract
Trigeminal neuropathic pain (TNP) is an intense pain condition characterized by hyperalgesia and allodynia; however, its neural mechanisms are not completely understood. Its management is complex, and studies that investigate its biochemical mechanisms are important for improving clinical approaches. This study aimed to evaluate the involvement of GABAergic, glutamatergic, and opioidergic systems and brain-derived neurotrophic factor (BDNF) levels in the TNP process in rats. TNP is induced by chronic constriction injury of the infraorbital nerve (CCI-ION). Nociceptive responses were evaluated using the facial von Frey test before and after the administration of GABAergic and opioidergic agonists and glutamatergic antagonists. The rats were divided into vehicle-treated control (C), sham-surgery (SS), and CCI-ION groups, and then subdivided into the vehicle (V)-treated SS-V and CCI-ION-V groups, SS-MK801 and CCI-ION-MK801, treated with the N-methyl-d-aspartate receptor selective antagonist MK801; SS-PB and CCI-ION-PB, treated with phenobarbital; SS-BZD and CCI-ION-BZD, treated with diazepam; SS-MOR and CCI-ION-MOR, treated with morphine. BDNF levels were evaluated in the cerebral cortex, brainstem, trigeminal ganglion, infraorbital branch of the trigeminal nerve, and serum. CCI-ION induced facial mechanical hyperalgesia. Phenobarbital and morphine reversed the hyperalgesia induced by CCI-ION, and the CCI-BZD group had an increased nociceptive threshold until 60 min. CCI-ION-GLU increased the nociceptive threshold at 60 min. Cerebral cortex and brainstem BDNF levels increased in the CCI-ION and SS groups. Only the CCI group presented high levels of BDNF in the trigeminal ganglion. Our data suggest the involvement of GABAergic, glutamatergic, and opioidergic systems and peripheral BDNF in the TNP process.
Collapse
Affiliation(s)
- Fabrício Finamor
- Nucleus of Pain Pharmacology and Neuromodulation. Hospital de Clínicas de Porto Alegre, RS, Brazil
| | - Vanessa Leal Scarabelot
- Nucleus of Pain Pharmacology and Neuromodulation. Hospital de Clínicas de Porto Alegre, RS, Brazil
| | - Liciane Fernandes Medeiros
- Nucleus of Pain Pharmacology and Neuromodulation. Hospital de Clínicas de Porto Alegre, RS, Brazil; Universidade La Salle, Canoas, RS, Brazil
| | - Dirson João Stein
- Nucleus of Pain Pharmacology and Neuromodulation. Hospital de Clínicas de Porto Alegre, RS, Brazil; School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Morgana Duarte da Silva
- Nucleus of Pain Pharmacology and Neuromodulation. Hospital de Clínicas de Porto Alegre, RS, Brazil
| | - Etiane Callai
- Nucleus of Pain Pharmacology and Neuromodulation. Hospital de Clínicas de Porto Alegre, RS, Brazil
| | - Wolnei Caumo
- Nucleus of Pain Pharmacology and Neuromodulation. Hospital de Clínicas de Porto Alegre, RS, Brazil; School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Andressa de Souza
- School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Iraci L S Torres
- Nucleus of Pain Pharmacology and Neuromodulation. Hospital de Clínicas de Porto Alegre, RS, Brazil; School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
10
|
Ulinastatin Alleviates Repetitive Ketamine Exposure-Evoked Cognitive Impairment in Adolescent Mice. Neural Plast 2022; 2022:6168284. [PMID: 36545238 PMCID: PMC9763019 DOI: 10.1155/2022/6168284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 10/13/2022] [Accepted: 11/01/2022] [Indexed: 12/14/2022] Open
Abstract
Ketamine (KET) is widely used for induction and maintenance of anesthesia, and long-term use is required for treatment of depression patients. Repeated use of KET is associated with mood and memory disorders. Ulinastatin (UTI), a urinary trypsin inhibitor, has been widely undertaken as an anti-inflammatory drug and proved to have neuroprotective effects. The aim of this work was to determine whether prophylactic use of UTI could attenuate KET-induced cognitive impairment. It was found that repetitive KET anesthesia cause cognitive and emotional disorders in adolescent mice in WMZ and OFT test, while UTI pretreatment reversed the poor performance compared to the AK group, and the platform finding time and center crossing time were obviously short in the CK+UTI group (P < 0.05). Our ELISA experiment results discovered that UTI pretreatment reduced the expression levels of IL-1β and IL-6 induced by CK anesthesia compared to AK (P < 0.05). In addition, UTI pretreatment protected the cognitive function by restraining the expression levels of Tau protein, Tau phospho-396 protein, and Aβ protein in the CK group compared to the AK group in Western blotting (P < 0.05). The results suggested that UTI could act as a new strategy to prevent the neurotoxicity of KET, revealing a significant neuroprotective effect of UTI.
Collapse
|
11
|
Mousavi S, Qiu H, Heinis FI, Abid MSR, Andrews MT, Checco JW. Short-Term Administration of Common Anesthetics Does Not Dramatically Change the Endogenous Peptide Profile in the Rat Pituitary. ACS Chem Neurosci 2022; 13:2888-2896. [PMID: 36126283 PMCID: PMC9547841 DOI: 10.1021/acschemneuro.2c00359] [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: 01/20/2023] Open
Abstract
Cell-cell signaling peptides (e.g., peptide hormones, neuropeptides) are among the largest class of cellular transmitters and regulate a variety of physiological processes. To identify and quantify the relative abundances of cell-cell signaling peptides in different physiological states, liquid chromatography-mass spectrometry-based peptidomics workflows are commonly utilized on freshly dissected tissues. In such animal experiments, the administration of general anesthetics is an important step for many research projects. However, acute anesthetic administration may rapidly change the measured abundance of transmitter molecules and metabolites, especially in the brain and endocrine system, which would confound experimental results. The aim of this study was to evaluate the effect of short-term (<5 min) anesthetic administration on the measured abundance of cell-cell signaling peptides, as evaluated by a typical peptidomics workflow. To accomplish this goal, we compared endogenous peptide abundances in the rat pituitary following administration of 5% isoflurane, 200 mg/kg sodium pentobarbital, or no anesthetic administration. Label-free peptidomics analysis demonstrated that acute use of isoflurane changed the levels of a small number of peptides, primarily degradation products of the hormone somatotropin, but did not influence the levels of most other peptide hormones. Acute use of sodium pentobarbital had negligible impact on the relative abundance of all measured peptides. Overall, our results suggest that anesthetics used in pituitary peptidomics studies do not dramatically confound observed results.
Collapse
Affiliation(s)
- Somayeh Mousavi
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, United States
| | - Haowen Qiu
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE 68588, United States
- The Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska-Lincoln, Lincoln, NE 68588, United States
| | - Frazer I. Heinis
- School of Natural Resources, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Md Shadman Ridwan Abid
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, United States
| | - Matthew T. Andrews
- School of Natural Resources, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - James W. Checco
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, United States
- The Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska-Lincoln, Lincoln, NE 68588, United States
| |
Collapse
|
12
|
Pavlovič A, Jakšová J, Kučerová Z, Špundová M, Rác M, Roudnický P, Mithöfer A. Diethyl ether anesthesia induces transient cytosolic [Ca 2+] increase, heat shock proteins, and heat stress tolerance of photosystem II in Arabidopsis. FRONTIERS IN PLANT SCIENCE 2022; 13:995001. [PMID: 36172556 PMCID: PMC9511054 DOI: 10.3389/fpls.2022.995001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/18/2022] [Indexed: 05/27/2023]
Abstract
General volatile anesthetic diethyl ether blocks sensation and responsive behavior not only in animals but also in plants. Here, using a combination of RNA-seq and proteomic LC-MS/MS analyses, we investigated the effect of anesthetic diethyl ether on gene expression and downstream consequences in plant Arabidopsis thaliana. Differential expression analyses revealed reprogramming of gene expression under anesthesia: 6,168 genes were upregulated, 6,310 genes were downregulated, while 9,914 genes were not affected in comparison with control plants. On the protein level, out of 5,150 proteins identified, 393 were significantly upregulated and 227 were significantly downregulated. Among the highest significantly downregulated processes in etherized plants were chlorophyll/tetrapyrrole biosynthesis and photosynthesis. However, measurements of chlorophyll a fluorescence did not show inhibition of electron transport through photosystem II. The most significantly upregulated process was the response to heat stress (mainly heat shock proteins, HSPs). Using transgenic A. thaliana expressing APOAEQUORIN, we showed transient increase of cytoplasmic calcium level [Ca2+]cyt in response to diethyl ether application. In addition, cell membrane permeability for ions also increased under anesthesia. The plants pre-treated with diethyl ether, and thus with induced HSPs, had increased tolerance of photosystem II to subsequent heat stress through the process known as cross-tolerance or priming. All these data indicate that diethyl ether anesthesia may partially mimic heat stress in plants through the effect on plasma membrane.
Collapse
Affiliation(s)
- Andrej Pavlovič
- Department of Biophysics, Faculty of Science, Palacký University, Olomouc, Czechia
| | - Jana Jakšová
- Department of Biophysics, Faculty of Science, Palacký University, Olomouc, Czechia
| | - Zuzana Kučerová
- Department of Biophysics, Faculty of Science, Palacký University, Olomouc, Czechia
| | - Martina Špundová
- Department of Biophysics, Faculty of Science, Palacký University, Olomouc, Czechia
| | - Marek Rác
- Department of Biophysics, Faculty of Science, Palacký University, Olomouc, Czechia
| | - Pavel Roudnický
- Central European Institute of Technology, Masaryk University, Brno, Czechia
| | - Axel Mithöfer
- Research Group Plant Defense Physiology, Max Planck Institute for Chemical Ecology, Jena, Germany
| |
Collapse
|
13
|
Adkins AM, Wellman LL, Sanford LD. Controllable and Uncontrollable Stress Differentially Impact Fear Conditioned Alterations in Sleep and Neuroimmune Signaling in Mice. Life (Basel) 2022; 12:1320. [PMID: 36143359 PMCID: PMC9506236 DOI: 10.3390/life12091320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 12/02/2022] Open
Abstract
Stress induces neuroinflammation and disrupts sleep, which together can promote a number of stress-related disorders. Fear memories associated with stress can resurface and reproduce symptoms. Our previous studies have demonstrated sleep outcomes can be modified by stressor controllability following stress and fear memory recall. However, it is unknown how stressor controllability alters neuroinflammatory signaling and its association with sleep following fear memory recall. Mice were implanted with telemetry transmitters and experienced escapable or inescapable footshock and then were re-exposed to the shuttlebox context one week later. Gene expression was assessed with Nanostring® panels using RNA extracted from the basolateral amygdala and hippocampus. Freezing and temperature were examined as behavioral measures of fear. Increased sleep after escapable stress was associated with a down-regulation in neuro-inflammatory and neuro-degenerative related genes, while decreased sleep after inescapable stress was associated with an up-regulation in these genes. Behavioral measures of fear were virtually identical. Sleep and neuroimmune responses appear to be integrated during fear conditioning and reproduced by fear memory recall. The established roles of disrupted sleep and neuroinflammation in stress-related disorders indicate that these differences may serve as informative indices of how fear memory can lead to psychopathology.
Collapse
Affiliation(s)
| | | | - Larry D. Sanford
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| |
Collapse
|
14
|
Robinson EJ, Lyne TC, Blaise BJ. Safety of general anaesthetics on the developing brain: are we there yet? BJA OPEN 2022; 2:100012. [PMID: 37588272 PMCID: PMC10430845 DOI: 10.1016/j.bjao.2022.100012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 04/11/2022] [Indexed: 08/18/2023]
Abstract
Thirty years ago, neurotoxicity induced by general anaesthetics in the developing brain of rodents was observed. In both laboratory-based and clinical studies, many conflicting results have been published over the years, with initial data confirming both histopathological and neurodevelopmental deleterious effects after exposure to general anaesthetics. In more recent years, animal studies using non-human primates and new human cohorts have identified some specific deleterious effects on neurocognition. A clearer pattern of neurotoxicity seems connected to exposure to repeated general anaesthesia. The biochemistry involved in this neurotoxicity has been explored, showing differential effects of anaesthetic drugs between the developing and developed brains. In this narrative review, we start with a comprehensive description of the initial concerning results that led to recommend that any non-essential surgery should be postponed after the age of 3 yr and that research into this subject should be stepped up. We then focus on the neurophysiology of the developing brain under general anaesthesia, explore the biochemistry of the observed neurotoxicity, before summarising the main scientific and clinical reports investigating this issue. We finally discuss the GAS trial, the importance of its results, and some potential limitations that should not undermine their clinical relevance. We finally suggest some key points that could be shared with parents, and a potential research path to investigate the biochemical effects of general anaesthesia, opening up perspectives to understand the neurocognitive effects of repetitive exposures, especially in at-risk children.
Collapse
Affiliation(s)
- Emily J. Robinson
- School of Population Health and Environmental Sciences, King's College London, London, UK
| | - Tom C. Lyne
- Center for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London, UK
| | - Benjamin J. Blaise
- Center for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London, UK
- Department of Paediatric Anaesthetics, Evelina London Children's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
15
|
Kim JJ, Sapio MR, Vazquez FA, Maric D, Loydpierson AJ, Ma W, Zarate CA, Iadarola MJ, Mannes AJ. Transcriptional Activation, Deactivation and Rebound Patterns in Cortex, Hippocampus and Amygdala in Response to Ketamine Infusion in Rats. Front Mol Neurosci 2022; 15:892345. [PMID: 35706427 PMCID: PMC9190438 DOI: 10.3389/fnmol.2022.892345] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
Ketamine, an N-methyl-D-aspartate (NMDA)-receptor antagonist, is a recently revitalized treatment for pain and depression, yet its actions at the molecular level remain incompletely defined. In this molecular-pharmacological investigation in the rat, we used short- and longer-term infusions of high dose ketamine to stimulate neuronal transcription processes. We hypothesized that a progressively stronger modulation of neuronal gene networks would occur over time in cortical and limbic pathways. A continuous intravenous administration paradigm for ketamine was developed in rat consisting of short (1 h) and long duration (10 h, and 10 h + 24 h recovery) infusions of anesthetic concentrations to activate or inhibit gene transcription in a pharmacokinetically controlled fashion. Transcription was measured by RNA-Seq in three brain regions: frontal cortex, hippocampus, and amygdala. Cellular level gene localization was performed with multiplex fluorescent in situ hybridization. Induction of a shared transcriptional regulatory network occurred within 1 h in all three brain regions consisting of (a) genes involved in stimulus-transcription factor coupling that are induced during altered synaptic activity (immediate early genes, IEGs, such as c-Fos, 9–12 significant genes per brain region, p < 0.01 per gene) and (b) the Nrf2 oxidative stress-antioxidant response pathway downstream from glutamate signaling (Nuclear Factor Erythroid-Derived 2-Like 2) containing 12–25 increasing genes (p < 0.01) per brain region. By 10 h of infusion, the acute results were further reinforced and consisted of more and stronger gene alterations reflecting a sustained and accentuated ketamine modulation of regional excitation and plasticity. At the cellular level, in situ hybridization localized up-regulation of the plasticity-associated gene Bdnf, and the transcription factors Nr4a1 and Fos, in cortical layers III and V. After 24 h recovery, we observed overshoot of transcriptional processes rather than a smooth return to homeostasis suggesting an oscillation of plasticity occurs during the transition to a new phase of neuronal regulation. These data elucidate critical molecular regulatory actions during and downstream of ketamine administration that may contribute to the unique drug actions of this anesthetic agent. These molecular investigations point to pathways linked to therapeutically useful attributes of ketamine.
Collapse
Affiliation(s)
- Jenny J. Kim
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Matthew R. Sapio
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Fernando A. Vazquez
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Amelia J. Loydpierson
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Wenting Ma
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Carlos A. Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Michael J. Iadarola
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Michael J. Iadarola, ,
| | - Andrew J. Mannes
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
16
|
Fu N, Zhu R, Zeng S, Li N, Zhang J. Effect of Anesthesia on Oligodendrocyte Development in the Brain. Front Syst Neurosci 2022; 16:848362. [PMID: 35664684 PMCID: PMC9158484 DOI: 10.3389/fnsys.2022.848362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
Oligodendrocytes (OLs) participate in the formation of myelin, promoting the propagation of action potentials, and disruption of their proliferation and differentiation leads to central nervous system (CNS) damage. As surgical techniques have advanced, there is an increasing number of children who undergo multiple procedures early in life, and recent experiments have demonstrated effects on brain development after a single or multiple anesthetics. An increasing number of clinical studies showing the effects of anesthetic drugs on the development of the nervous system may mainly reside in the connections between neurons, where myelin development will receive more research attention. In this article, we review the relationship between anesthesia exposure and the brain and OLs, provide new insights into the development of the relationship between anesthesia exposure and OLs, and provide a theoretical basis for clinical prevention of neurodevelopmental risks of general anesthesia drugs.
Collapse
|
17
|
Miranda A, Bertoglio D, Stroobants S, Staelens S, Verhaeghe J. Translation of Preclinical PET Imaging Findings: Challenges and Motion Correction to Overcome the Confounding Effect of Anesthetics. Front Med (Lausanne) 2021; 8:753977. [PMID: 34746189 PMCID: PMC8569248 DOI: 10.3389/fmed.2021.753977] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/27/2021] [Indexed: 11/13/2022] Open
Abstract
Preclinical brain positron emission tomography (PET) in animals is performed using anesthesia to avoid movement during the PET scan. In contrast, brain PET scans in humans are typically performed in the awake subject. Anesthesia is therefore one of the principal limitations in the translation of preclinical brain PET to the clinic. This review summarizes the available literature supporting the confounding effect of anesthesia on several PET tracers for neuroscience in preclinical small animal scans. In a second part, we present the state-of-the-art methodologies to circumvent this limitation to increase the translational significance of preclinical research, with an emphasis on motion correction methods. Several motion tracking systems compatible with preclinical scanners have been developed, each one with its advantages and limitations. These systems and the novel experimental setups they can bring to preclinical brain PET research are reviewed here. While technical advances have been made in this field, and practical implementations have been demonstrated, the technique should become more readily available to research centers to allow for a wider adoption of the motion correction technique for brain research.
Collapse
Affiliation(s)
- Alan Miranda
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Daniele Bertoglio
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Sigrid Stroobants
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
- University Hospital Antwerp, Antwerp, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Jeroen Verhaeghe
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
18
|
Gu Q, Kanungo J. Effect of ketamine on gene expression in zebrafish embryos. J Appl Toxicol 2021; 41:2083-2089. [PMID: 34002392 DOI: 10.1002/jat.4199] [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: 04/13/2021] [Revised: 05/03/2021] [Accepted: 05/03/2021] [Indexed: 01/21/2023]
Abstract
Ketamine is an N-methyl-D-aspartate (NMDA) receptor antagonist. Used as an anesthetic, potential neurotoxic and cardiotoxic effects of ketamine in animal models have been reported. The underlying mechanisms of ketamine-induced toxicity are not clear. The zebrafish is an ideal model for toxicity assays because of its predictive capability in chemical testing, which compares well with that of mammalian models. To gain insight into potential mechanisms of ketamine effects, we performed real-time quantitative polymerase chain reaction-based gene expression array analyses. Gene expression analysis was conducted for multiple genes (a total of 84) related to 10 major signaling pathways including the transforming growth factor β (TGFβ), Wingless and Int-1 (Wnt), nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB), Janus kinase/signal transducers and activators of transcription (JAK/STAT), p53, Notch, Hedgehog, peroxisome proliferator-activated receptor (PPAR), oxidative stress, and hypoxia pathways. Our results show that ketamine altered the expression of specific genes related to hypoxia, p53, Wnt, Notch, TGFβ, PPAR, and oxidative stress pathways. Thus, we can further focus on these specific pathways to elucidate the mechanisms by which ketamine elicits a toxic response.
Collapse
Affiliation(s)
- Qiang Gu
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Jyotshna Kanungo
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| |
Collapse
|
19
|
Nunes-Freitas AL, Soni N, Polli FS, Kohlmeier KA. Prenatal exposure to nicotine in mice is associated with alterations in development and cellular and synaptic effects of alcohol in a brainstem arousal nucleus. Neurotoxicol Teratol 2021; 87:106980. [PMID: 33838245 DOI: 10.1016/j.ntt.2021.106980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/22/2021] [Accepted: 04/04/2021] [Indexed: 02/07/2023]
Abstract
Using drugs of abuse while pregnant has tremendous negative consequences for the offspring, including an enhanced risk for substance use disorder (SUD). This vulnerability suggests that gestational exposure to drugs alters the developmental trajectory of neurons important in SUD processes, which could lead to later life changes in responsiveness to motivationally salient stimuli. The laterodorsal tegmentum (LDT) gates the behaviorally relevant firing pattern signaling stimuli saliency in mesoaccumbal circuits. Accordingly, any alterations in LDT functionality could alter output, and play a role in negative outcomes on motivated behavior associated with early-life nicotine exposure. Therefore, we investigated whether prenatal exposure to nicotine (PNE), which is a known teratogen, altered responsiveness of LDT neurons to alcohol by conducting electrophysiology in brain slices. Alcohol induced an outward current in control LDT cells, which was not seen in PNE LDT neurons. The frequency of mEPSCs was significantly decreased by alcohol in LDT PNE cells and accompanied by a decrease in action potential frequency, which were actions not seen in controls. Changes in baseline activity of PNE LDT cells were also observed. In summary, PNE LDT neurons showed alterations in baseline activity and membrane and synaptic responses to postnatal exposures to alcohol. The differences in PNE baseline activity and alcohol responses likely lead to differential output from the LDT to mesoaccumbal targets that could play a role in biasing coding of relevant stimuli, which could participate in the enhanced proclivity for development of SUD in those exposed during gestation to nicotine.
Collapse
Affiliation(s)
- André Luiz Nunes-Freitas
- Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark; Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Neeraj Soni
- Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Filip S Polli
- Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Kristi A Kohlmeier
- Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark.
| |
Collapse
|
20
|
Yang Y, Yi J, Pan M, Hu B, Duan H. Edaravone Alleviated Propofol-Induced Neurotoxicity in Developing Hippocampus by mBDNF/TrkB/PI3K Pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:1409-1422. [PMID: 33833500 PMCID: PMC8020057 DOI: 10.2147/dddt.s294557] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 03/09/2021] [Indexed: 11/25/2022]
Abstract
Background To investigate the neuroprotective effect of edaravone on excessive-dose propofol-induced neurotoxicity in the hippocampus of newborn rats and HT22 cells. Methods Cell proliferation was investigated by assessing ki67 expression in the neural stem of the hippocampus of newborn rats and by cell counting kit-8 (CCK8) assay in HT22 cells. Cell apoptosis was assessed in vivo by caspase 3 detection in Western blots and measurement of apoptosis in neurons and glial cells by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. Apoptosis was analyzed by flow cytometry in HT22 cells. The Morris water maze was used to evaluate the long-term learning and memory ability of rats. Inflammatory factors were detected by enzyme-linked immunosorbent assay (ELISA). The expression of mBDNF/TrkB/PI3K pathway-related proteins was detected by Western blot and quantitative reverse transcription-polymerase chain reaction (q-RT PCR). Results In neonatal rat hippocampus and HT22 cells, edaravone increased cell proliferation and decreased cell apoptosis after excessive propofol-induced neurotoxicity. In addition, the levels of proinflammatory factors interleukin (IL)-6 and tumor necrosis factor (TNF)-α were reduced by edaravone pretreatment. The use of the tropomyosin receptor kinase B (TrkB) antagonist ANA-12 and TrkB agonist 7,8DHF with propofol groups showed that edaravone mitigated excessive propofol-induced neurotoxicity through the mature brain-derived neurotrophic factor (mBDNF)/TrkB/phosphoinositide 3-kinase (PI3K) pathway. However, the current dose of propofol did not significantly affect long-term learning and memory in rats. Conclusion Edaravone pretreatment ameliorated propofol-induced proliferation inhibition, neuroapoptosis, and neural inflammation by activating the mBDNF/TrkB/PI3K pathway.
Collapse
Affiliation(s)
- Yangliang Yang
- Department of Anesthesiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, People's Republic of China
| | - Jing Yi
- Department of Anesthesiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, People's Republic of China
| | - Mengzhi Pan
- Department of Anesthesiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, People's Republic of China
| | - Baoji Hu
- Department of Anesthesiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, People's Republic of China
| | - Hongwei Duan
- Department of Anesthesiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, People's Republic of China
| |
Collapse
|
21
|
Lisek M, Zylinska L, Boczek T. Ketamine and Calcium Signaling-A Crosstalk for Neuronal Physiology and Pathology. Int J Mol Sci 2020; 21:ijms21218410. [PMID: 33182497 PMCID: PMC7665128 DOI: 10.3390/ijms21218410] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/31/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022] Open
Abstract
Ketamine is a non-competitive antagonist of NMDA (N-methyl-D-aspartate) receptor, which has been in clinical practice for over a half century. Despite recent data suggesting its harmful side effects, such as neuronal loss, synapse dysfunction or disturbed neural network formation, the drug is still applied in veterinary medicine and specialist anesthesia. Several lines of evidence indicate that structural and functional abnormalities in the nervous system caused by ketamine are crosslinked with the imbalanced activity of multiple Ca2+-regulated signaling pathways. Due to its ubiquitous nature, Ca2+ is also frequently located in the center of ketamine action, although the precise mechanisms underlying drug’s negative or therapeutic properties remain mysterious for the large part. This review seeks to delineate the relationship between ketamine-triggered imbalance in Ca2+ homeostasis and functional consequences for downstream processes regulating key aspects of neuronal function.
Collapse
|