1
|
Wellington NJ, Boųcas AP, Lagopoulos J, Quigley BL, Kuballa AV. Molecular pathways of ketamine: A systematic review of immediate and sustained effects on PTSD. Psychopharmacology (Berl) 2025; 242:1197-1243. [PMID: 40097854 DOI: 10.1007/s00213-025-06756-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025]
Abstract
RATIONALE Existing studies predominantly focus on the molecular and neurobiological mechanisms underlying Ketamine's acute treatment effects on post-traumatic stress disorder (PTSD). This emphasis has largely overlooked its sustained therapeutic effects, which hold significant potential for the development of targeted interventions. OBJECTIVES This systematic review examines the pharmacokinetic and pharmacodynamic effects of ketamine on PTSD, differentiating between immediate and sustained molecular effects. METHOD A comprehensive search across databases (Web of Science, Scopus, Global Health, PubMed) and grey literature yielded 317 articles, where 29 studies met the inclusion criteria. These studies included preclinical models and clinical trials, through neurotransmitter regulation, gene expression, synaptic plasticity, and neural pathways (PROSPERO ID: CRD42024582874). RESULTS We found accumulating evidence that the immediate effects of ketamine, which involve changes in GABA, glutamate, and glutamine levels, trigger the re-regulation of BDNF, enhancing synaptic plasticity via pathways such as TrkB and PSD-95. Other molecular influences also include c-Fos, GSK-3, HDAC, HCN1, and the modulation of hormones like CHR and ACTH, alongside immune responses (IL-6, IL-1β, TNF-α). Sustained effects arise from neurotransmitter remodulations and involve prolonged changes in gene expression. These include mTOR-mediated BDNF expression, alterations in GSK-3β, FkBP5, GFAP, ERK phosphorylation, and epigenetic modifications (DNMT3, MeCP2, H3K27me3, mir-132, mir-206, HDAC). CONCLUSION These molecular changes promote long-term synaptic stability and re-regulation in key brain regions, contributing to prolonged therapeutic benefits. Understanding the sustained molecular and epigenetic mechanisms behind ketamine's effects is critical for developing safe and effective personalised treatments, potentially leading to more effective recovery.
Collapse
Affiliation(s)
- Nathan J Wellington
- National PTSD Research Centre, Thompson Institute, University of the Sunshine Coast (UniSC), Birtinya, QLD, Australia.
- School of Health, UniSC, Sippy Downs, QLD, Australia.
- Centre for Bioinnovation, UniSC, Sippy Downs, QLD, Australia.
- Sunshine Coast Hospital and Health Service, Sunshine Coast Health Institute, Birtinya, QLD, Australia.
| | - Ana P Boųcas
- National PTSD Research Centre, Thompson Institute, University of the Sunshine Coast (UniSC), Birtinya, QLD, Australia
| | - Jim Lagopoulos
- Thompson Brain and Mind Healthcare, Maroochydore, QLD, Australia
| | - Bonnie L Quigley
- National PTSD Research Centre, Thompson Institute, University of the Sunshine Coast (UniSC), Birtinya, QLD, Australia
- Centre for Bioinnovation, UniSC, Sippy Downs, QLD, Australia
- Sunshine Coast Hospital and Health Service, Sunshine Coast Health Institute, Birtinya, QLD, Australia
| | - Anna V Kuballa
- School of Health, UniSC, Sippy Downs, QLD, Australia
- Centre for Bioinnovation, UniSC, Sippy Downs, QLD, Australia
- Sunshine Coast Hospital and Health Service, Sunshine Coast Health Institute, Birtinya, QLD, Australia
| |
Collapse
|
2
|
Faustino Martins AC, Badenoch B, da Silva Gomes R. Insights for the Next Generation of Ketamine for the Treatment of Depressive Disorder. J Med Chem 2025; 68:944-952. [PMID: 39757458 PMCID: PMC12077806 DOI: 10.1021/acs.jmedchem.4c02467] [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: 01/07/2025]
Abstract
Treatment-resistant depression responds quickly to ketamine. As an N-methyl-d-aspartate receptor (NMDAR) antagonist, ketamine may affect prefrontal cortex (PFC) neurons. Recent investigations reveal that the (R)-enantiomer is the most effective and least abuseable antidepressant. The Food and Drug Administration approves only the (S)-enantiomer for medical usage. (2R,6R)-Hydroxynorketamine (HNK) inhibits mGlu2, linked to a Gi, in presynaptic glutamatergic neurons, increasing brain-derived neurotrophic factor (BDNF) release, which autocrinely activates Tropomyosin receptor kinase B (TrkB) and promotes synaptogenesis. Ketamine, originally an anesthetic, has garnered attention for its many pharmacological effects, including its potential as a rapid-acting antidepressant and recreational use. In this Perspective, we explore the synthesis, pharmacology, metabolism, and effects of ketamine and its metabolites in animal and human studies to explain the difference in the biological activity between the enantiomers.
Collapse
Affiliation(s)
- Allana Cristina Faustino Martins
- Department of Pharmaceutical Sciences, College of Health and Human Sciences, North Dakota State University, Fargo, ND, 58105, United States
| | - Bretton Badenoch
- Department of Pharmaceutical Sciences, College of Health and Human Sciences, North Dakota State University, Fargo, ND, 58105, United States
| | - Roberto da Silva Gomes
- Department of Pharmaceutical Sciences, College of Health and Human Sciences, North Dakota State University, Fargo, ND, 58105, United States
| |
Collapse
|
3
|
Ma S, Xu CC, Dong YC, Li CX, Shang YZ. Scutellaria Baicalensis Georgi Stem and Leaf Flavonoids Ameliorate the Learning and Memory Impairment in Rats Induced by Okadaic Acid. Comb Chem High Throughput Screen 2025; 28:263-277. [PMID: 38204252 DOI: 10.2174/0113862073265060231127054325] [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: 06/20/2023] [Revised: 10/05/2023] [Accepted: 10/17/2023] [Indexed: 01/12/2024]
Abstract
AIM The objective of this study is to explore the impact and underlying mechanism of S. baicalensis Georgi stem and leaf flavonoids (SSFs) on cognitive impairment caused by intracerebroventricular injection of okadaic acid (OA) in rats. METHODS An experimental model of Alzheimer's disease (AD) was induced in rats by intracerebroventricular injection of OA, resulting in memory impairment. The Morris water maze test was employed to confirm the successful establishment of the memory impairment model. The rats that exhibited significant memory impairment were randomly divided into different groups, including a model group, three SSFs dose groups (25, 50, and 100 mg/kg), and a positive control group treated with Ginkgo biloba tablets (GLT) at a dose of 200 mg/kg. To evaluate the learning and memory abilities of the rats, the Morris water maze test was conducted. Hematoxylin-eosin (HE) staining was used to observe any morphological changes in neurons. Immunohistochemistry (IHC) was performed to measure the expression of choline acetyltransferase (ChAT) protein. Western blotting (WB) was utilized to assess the phosphorylation levels of tau protein at Ser262 and Ser396. The activities of inducible nitric oxide synthase (iNOS) and constitutive nitric oxide synthase (cNOS) were quantified using ultraviolet spectrophotometry. The levels of inflammatory factors, including interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6), were measured using ELISA. RESULTS In rats, the administration of OA via intracerebroventricular injection resulted in cognitive impairment, neuropathological changes, and alterations in protein expression and activity levels. Specifically, the protein expression of ChAT was significantly reduced (P<0.01), while the phosphorylation levels of tau protein at Ser262 and Ser396 were significantly increased (P<0.01). Moreover, iNOS activity in the hippocampus and cerebral cortex exhibited a significant increase (P<0.01), whereas cNOS activity showed a decrease (P<0.05). Furthermore, the levels of IL-1β and TNF-α in the cerebral cortex were elevated (P<0.01), while the level of IL-6 was decreased (P<0.05). The administration of three doses of SSFs and GLT to rats exhibited varying degrees of improvement in the aforementioned pathological alterations induced by OA. CONCLUSION SSFs demonstrated the ability to enhance cognitive function and mitigate memory deficits in rats following intracerebroventricular injection of OA. This beneficial effect may be attributed to the modulation of ChAT protein expression, tau hyperphosphorylation, NOS activity, and inflammatory cytokine levels by SSFs.
Collapse
Affiliation(s)
- Shuai Ma
- Institute of Traditional Chinese Medicine, Chengde Medical University / Hebei Province Key Research Office of Traditional Chinese Medicine Against Dementia / Hebei Province Key Laboratory of Traditional Chinese Medicine Research and Development / Hebei Key Laboratory of Nerve Injury and Repair, Anyuan Road, Chengde, 067000, P.R. China
| | - Cong-Cong Xu
- Institute of Traditional Chinese Medicine, Chengde Medical University / Hebei Province Key Research Office of Traditional Chinese Medicine Against Dementia / Hebei Province Key Laboratory of Traditional Chinese Medicine Research and Development / Hebei Key Laboratory of Nerve Injury and Repair, Anyuan Road, Chengde, 067000, P.R. China
| | - Yong-Cai Dong
- Institute of Traditional Chinese Medicine, Chengde Medical University / Hebei Province Key Research Office of Traditional Chinese Medicine Against Dementia / Hebei Province Key Laboratory of Traditional Chinese Medicine Research and Development / Hebei Key Laboratory of Nerve Injury and Repair, Anyuan Road, Chengde, 067000, P.R. China
| | - Cai-Xia Li
- The Fourth Hospital of Shijiazhuang, Shijiazhuang, 050011, China
| | - Ya-Zhen Shang
- Institute of Traditional Chinese Medicine, Chengde Medical University / Hebei Province Key Research Office of Traditional Chinese Medicine Against Dementia / Hebei Province Key Laboratory of Traditional Chinese Medicine Research and Development / Hebei Key Laboratory of Nerve Injury and Repair, Anyuan Road, Chengde, 067000, P.R. China
| |
Collapse
|
4
|
Choi H, Hwang W. Anesthetic Approaches and Their Impact on Cancer Recurrence and Metastasis: A Comprehensive Review. Cancers (Basel) 2024; 16:4269. [PMID: 39766169 PMCID: PMC11674873 DOI: 10.3390/cancers16244269] [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: 10/21/2024] [Revised: 11/10/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Cancer recurrence and metastasis remain critical challenges following surgical resection, influenced by complex perioperative mechanisms. This review explores how surgical stress triggers systemic changes, such as neuroendocrine responses, immune suppression, and inflammation, which promote the dissemination of residual cancer cells and circulating tumor cells. Key mechanisms, such as epithelial-mesenchymal transition and angiogenesis, further enhance metastasis, while hypoxia-inducible factors and inflammatory responses create a microenvironment conducive to tumor progression. Anesthetic agents and techniques modulate these mechanisms in distinct ways. Inhaled anesthetics, such as sevoflurane, may suppress immune function by increasing catecholamines and cytokines, thereby promoting cancer progression. In contrast, propofol-based total intravenous anesthesia mitigates stress responses and preserves natural killer cell activity, supporting immune function. Opioids suppress immune surveillance and promote angiogenesis through the activation of the mu-opioid receptor. Opioid-sparing strategies using NSAIDs show potential in preserving immune function and reducing recurrence risk. Regional anesthesia offers benefits by reducing systemic stress and immune suppression, though the clinical outcomes remain inconsistent. Additionally, dexmedetomidine and ketamine exhibit dual effects, both enhancing and inhibiting tumor progression depending on the dosage and context. This review emphasizes the importance of individualized anesthetic strategies to optimize long-term cancer outcomes. While retrospective studies suggest potential benefits of propofol-based total intravenous anesthesia and regional anesthesia, further large-scale trials are essential to establish the definitive role of anesthetic management in cancer recurrence and survival.
Collapse
Affiliation(s)
| | - Wonjung Hwang
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| |
Collapse
|
5
|
Huh J, Hwang W. The Role of Anesthetic Management in Lung Cancer Recurrence and Metastasis: A Comprehensive Review. J Clin Med 2024; 13:6681. [PMID: 39597826 PMCID: PMC11594908 DOI: 10.3390/jcm13226681] [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: 10/14/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Lung cancer remains a leading cause of cancer-related mortality worldwide. Although surgical treatment is a primary approach, residual cancer cells and surgery-induced pathophysiological changes may promote cancer recurrence and metastasis. Anesthetic agents and techniques have recently been shown to potentially impact these processes by modulating surgical stress responses, immune function, inflammatory pathways, and the tumor microenvironment. Anesthetics can influence immune-modulating cytokines, induce pro-inflammatory factors such as HIF-1α, and alter natural-killer cell activity, affecting cancer cell survival and spread. Preclinical studies suggest volatile anesthetics may promote tumor progression by triggering pro-inflammatory signaling, while propofol shows potential antitumor properties through immune-preserving effects and reductions in IL-6 and other inflammatory markers. Additionally, opioids are known to suppress immune responses and stimulate pathways that may support cancer cell proliferation, whereas regional anesthesia may reduce these risks by decreasing the need for systemic opioids and volatile agents. Despite these findings, clinical data remain inconclusive, with studies showing mixed outcomes across patient populations. Current clinical trials, including comparisons of volatile agents with propofol-based total intravenous anesthesia, aim to provide clarity but highlight the need for further investigation. Large-scale, well-designed studies are essential to validate the true impact of anesthetic choice on cancer recurrence and to optimize perioperative strategies that support long-term oncologic outcomes for lung cancer patients.
Collapse
Affiliation(s)
| | - Wonjung Hwang
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| |
Collapse
|
6
|
Wang P, Jiang L, Hu J, Jiang Z, Zhang Y, Chen C, Lin Y, Su M, Wang X, Liao L. The amino acid metabolism pathway of peripheral T lymphocytes and ketamine-induced schizophrenia-like phenotype. J Psychiatry Neurosci 2024; 49:E413-E426. [PMID: 39626901 PMCID: PMC11633891 DOI: 10.1503/jpn-240038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND The intricate interplay between peripheral adaptive immune cells and the central nervous system (CNS) has garnered increasing recognition. Given that alterations in cell quantities often translate into modifications in metabolite profiles and that these metabolic changes can potentially traverse the bloodstream and enter the CNS, thereby modulating the progression of mental illnesses, we sought to explore the metabolic profiles of peripheral immune cells in a ketamine-treated mouse model of schizophrenia. METHODS We used flow cytometry to scrutinize the alterations in peripheral adaptive immune cells in a ketamine-induced schizophrenia mouse model. Subsequently, we implemented an untargeted metabolomic approach with ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) to detect the metabolite profiles of peripheral abnormal lymphocytes and identify differential metabolites present in plasma. We then employed targeted metabolomics using UPLC-MS/MS to quantify the common differential metabolites detected in mouse plasma. RESULTS Flow cytometry analysis detected a notable increase in the count of peripheral CD3+ T cells in a ketamine-induced schizophrenia mouse model. Subsequent untargeted metabolomics analysis revealed that the amino acid metabolism pathway underwent substantial alterations. A detailed quantification of 22 amino acid profiles in the peripheral plasma indicated significant elevation in the levels of glycine, alanine, asparagine, and aspartic acid. LIMITATIONS Our ongoing research has yet to conclusively identify the precise amino acid metabolism pathway that serves as the pivotal factor in the manifestation of the schizophrenia-like phenotype induced by ketamine. CONCLUSION The peripheral amino acid metabolism pathway is involved in the ketamine-induced schizophrenia-like phenotype. The metabolic profile of peripheral immune cells could provide accurate biomarkers for the diagnosis and treatment of psychiatric diseases.
Collapse
Affiliation(s)
- Peipei Wang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Linzhi Jiang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Junmei Hu
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Zihan Jiang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Yu Zhang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Congliang Chen
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Yanchen Lin
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Mi Su
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Xia Wang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Linchuan Liao
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Afshar Ghahremani S, Raisi A, Minaei Beirami S, Kahroba H, Mardani M, Dezfoulian O, Tarhriz V. Curcumin alleviates inflammatory effects of ketamine anesthesia in postnatal rats. VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2024; 15:473-480. [PMID: 39564472 PMCID: PMC11571040 DOI: 10.30466/vrf.2024.2018359.4107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/05/2024] [Indexed: 11/21/2024]
Abstract
Curcumin has been employed in traditional medicine for over a millennium to treat various ailments, and its global use is now widespread. Chinese medicine relies heavily on curcumin as a primary element and uses it to cure infectious diseases, skin disorders, depression, and stress. It has cardioprotective, neuroprotective, and anti-diabetic properties, as well as pharmacological effects on disorders like type II diabetes, atherosclerosis, and human immunodeficiency virus replication. The anti-cancer activity of curcumin has been studied extensively with notable improvements in gastrointestinal, melanoma, urogenital, breast, and lung malignancies. We investigated the anti-inflammatory effects of curcumin on expression of tumor necrosis factor (TNF)-α, c-Fos, and interleukin (IL)-6 genes in brain and liver tissue owing to the effects of ketamine anesthesia on postnatal rats. The thalamic and hepatic tissues were collected without anesthesia, immediately after anesthesia, and 4 and 12 hr after anesthesia in control and curcumin treated postnatal rats. The results showed that glucose, triglyceride, high- and low-density lipoprotein levels were lowered with curcumin treatment. We also found that ketamine increased c-Fos and inflammatory cytokines like TNF-α and IL-6, all of which contribute to inflammation. Brain and liver immunohistochemistry studies confirmed the real-time polymerase chain reaction findings. Curcumin injections alone may be effective in decreasing ketamine-induced inflammation in both brain and liver tissues.
Collapse
Affiliation(s)
- Soroush Afshar Ghahremani
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| | - Abbas Raisi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| | - Sohrab Minaei Beirami
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Houman Kahroba
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahnaz Mardani
- Department of Health and Nutrition, Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Omid Dezfoulian
- Department of Pathobiology, Faculty of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| | - Vahideh Tarhriz
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, USA
| |
Collapse
|
8
|
Wang S, Yu X, Cheng L, Ren W, Wen G, Wu X, Lou H, Ren X, Lu L, Hermenean A, Yao J, Li B, Lu Y, Wu X. Dexmedetomidine improves the circulatory dysfunction of the glymphatic system induced by sevoflurane through the PI3K/AKT/ΔFosB/AQP4 pathway in young mice. Cell Death Dis 2024; 15:448. [PMID: 38918408 PMCID: PMC11199640 DOI: 10.1038/s41419-024-06845-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024]
Abstract
Multiple sevoflurane exposures may damage the developing brain. The neuroprotective function of dexmedetomidine has been widely confirmed in animal experiments and human studies. However, the effect of dexmedetomidine on the glymphatic system has not been clearly studied. We hypothesized that dexmedetomidine could alleviate sevoflurane-induced circulatory dysfunction of the glymphatic system in young mice. Six-day-old C57BL/6 mice were exposed to 3% sevoflurane for 2 h daily, continuously for 3 days. Intraperitoneal injection of either normal saline or dexmedetomidine was administered before every anaesthesia. Meanwhile the circulatory function of glymphatic system was detected by tracer injection at P8 and P32. On P30-P32, behavior tests including open field test, novel object recognition test, and Y-maze test were conducted. Primary astrocyte cultures were established and treated with the PI3K activator 740Y-P, dexmedetomidine, and small interfering RNA (siRNA) to silence ΔFosB. We propose for the first time that multiple exposure to sevoflurane induces circulatory dysfunction of the glymphatic system in young mice. Dexmedetomidine improves the circulatory capacity of the glymphatic system in young mice following repeated exposure to sevoflurane through the PI3K/AKT/ΔFosB/AQP4 signaling pathway, and enhances their long-term learning and working memory abilities.
Collapse
Affiliation(s)
- Shuying Wang
- School of Forensic Medicine, China Medical University, Shenyang, China
- Department of Anaesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Xiaojin Yu
- School of Forensic Medicine, China Medical University, Shenyang, China
- Department of Anaesthesiology, Affiliated Shengjing Hospital of China Medical University, Shenyang, China
| | - Lili Cheng
- School of Forensic Medicine, China Medical University, Shenyang, China
- Department of Anaesthesiology, Affiliated Shengjing Hospital of China Medical University, Shenyang, China
| | - Weishu Ren
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Gehua Wen
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Xue Wu
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Haoyang Lou
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Xinghua Ren
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Lei Lu
- Department of pediatrics Neonatology, University of Chicago, Chicago, IL, 60615, USA
| | - Anca Hermenean
- Faculty of Medicine, Vasile Goldis Western University of Arad, Arad, Romania
| | - Jun Yao
- School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Center of Forensic Investigation, Shenyang, China
| | - Baoman Li
- School of Forensic Medicine, China Medical University, Shenyang, China.
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China.
- China Medical University Center of Forensic Investigation, Shenyang, China.
| | - Yan Lu
- Key Laboratory of Health Ministry in Congenital Malformation, Affiliated Shengjing Hospital of China Medical University, Shenyang, China.
| | - Xu Wu
- School of Forensic Medicine, China Medical University, Shenyang, China.
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China.
- China Medical University Center of Forensic Investigation, Shenyang, China.
| |
Collapse
|
9
|
Yang Y, Hang W, Li J, Liu T, Hu Y, Fang F, Yan D, McQuillan PM, Wang M, Hu Z. Effect of General Anesthetic Agents on Microglia. Aging Dis 2024; 15:1308-1328. [PMID: 37962460 PMCID: PMC11081156 DOI: 10.14336/ad.2023.1108] [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: 09/07/2023] [Accepted: 11/08/2023] [Indexed: 11/15/2023] Open
Abstract
The effects of general anesthetic agents (GAAs) on microglia and their potential neurotoxicity have attracted the attention of neuroscientists. Microglia play important roles in the inflammatory process and in neuromodulation of the central nervous system. Microglia-mediated neuroinflammation is a key mechanism of neurocognitive dysfunction during the perioperative period. Microglial activation by GAAs induces anti-inflammatory and pro-inflammatory effects in microglia, suggesting that GAAs play a dual role in the mechanism of postoperative cognitive dysfunction. Understanding of the mechanisms by which GAAs regulate microglia may help to reduce the incidence of postoperative adverse effects. Here, we review the actions of GAAs on microglia and the consequent changes in microglial function. We summarize clinical and animal studies associating microglia with general anesthesia and describe how GAAs interact with neurons via microglia to further explore the mechanisms of action of GAAs in the nervous system.
Collapse
Affiliation(s)
- Yanchang Yang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Wenxin Hang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jun Li
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Anesthesiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China.
| | - Tiantian Liu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Anesthesiology, Ningbo Women and Children's Hospital, Ningbo, China.
| | - Yuhan Hu
- Cell Biology Department, Yale University, New Haven, CT, USA.
| | - Fuquan Fang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Dandan Yan
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Patrick M. McQuillan
- Department of Anesthesiology, Penn State Hershey Medical Center, Penn State College of Medicine, Hershey, PA, USA.
| | - Mi Wang
- Department of Anesthesiology, The First Affiliated 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
|
10
|
Megha KB, Mohanan PV. Cellular consequences triggered by ketamine on exposure to human glioblastoma epithelial (LN-229) cells. J Biochem Mol Toxicol 2023; 37:e23484. [PMID: 37515540 DOI: 10.1002/jbt.23484] [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: 02/13/2023] [Revised: 06/09/2023] [Accepted: 07/20/2023] [Indexed: 07/31/2023]
Abstract
Ketamine is generally a noncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist that interrelates with various other receptors, contributing to a wide range of actions. They are mainly approved as a general anesthetic, but a low dose of ketamine is applied for pain management, depression, and as analgesics. However, there is a significant concern regarding the long-term usage as antidepressants and as an abused drug. The study mainly aims to exhibit the possible long-term side effects of ketamine as an antidepressant and in recreational users. The study explores the in vitro cytotoxicity revealed on LN-229 cells in a dose-dependent manner. According to the cell viability assays, there is a dose-dependent response toward ketamine. Morphological and nuclear integrity was changed on exposure and assessed using Giemsa, Rhodamine phalloidin, 4',6-diamidino-2-phenylindole (DAPI), and Acridine orange staining. The apoptotic cell death marked by nuclear condensation, Lactate dehydrogenase leakage, pro-inflammatory cytokine (interleukin [IL]-β) release, and inhibition of cell migration was observed. The study highlights the importance of nonanesthetic usage of ketamine, which can lead to severe adverse side effects on long-term exposure rather than a single exposure as an anesthetic agent.
Collapse
Affiliation(s)
- Kizhakkepurakkal B Megha
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, India
| | - Parayanthala V Mohanan
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, India
| |
Collapse
|
11
|
Layton R, Layton D, Beggs D, Fisher A, Mansell P, Stanger KJ. The impact of stress and anesthesia on animal models of infectious disease. Front Vet Sci 2023; 10:1086003. [PMID: 36816193 PMCID: PMC9933909 DOI: 10.3389/fvets.2023.1086003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/09/2023] [Indexed: 02/05/2023] Open
Abstract
Stress and general anesthesia have an impact on the functional response of the organism due to the detrimental effects on cardiovascular, immunological, and metabolic function, which could limit the organism's response to an infectious event. Animal studies have formed an essential step in understanding and mitigating infectious diseases, as the complexities of physiology and immunity cannot yet be replicated in vivo. Using animals in research continues to come under increasing societal scrutiny, and it is therefore crucial that the welfare of animals used in disease research is optimized to meet both societal expectations and improve scientific outcomes. Everyday management and procedures in animal studies are known to cause stress, which can not only cause poorer welfare outcomes, but also introduces variables in disease studies. Whilst general anesthesia is necessary at times to reduce stress and enhance animal welfare in disease research, evidence of physiological and immunological disruption caused by general anesthesia is increasing. To better understand and quantify the effects of stress and anesthesia on disease study and welfare outcomes, utilizing the most appropriate animal monitoring strategies is imperative. This article aims to analyze recent scientific evidence about the impact of stress and anesthesia as uncontrolled variables, as well as reviewing monitoring strategies and technologies in animal models during infectious diseases.
Collapse
Affiliation(s)
- Rachel Layton
- Australian Centre for Disease Preparedness, CSIRO, Geelong, VIC, Australia,*Correspondence: Rachel Layton ✉
| | - Daniel Layton
- Australian Centre for Disease Preparedness, CSIRO, Geelong, VIC, Australia
| | - David Beggs
- Faculty of Veterinary and Agricultural Sciences, Melbourne Veterinary School, University of Melbourne, Melbourne, VIC, Australia
| | - Andrew Fisher
- Faculty of Veterinary and Agricultural Sciences, Melbourne Veterinary School, University of Melbourne, Melbourne, VIC, Australia
| | - Peter Mansell
- Faculty of Veterinary and Agricultural Sciences, Melbourne Veterinary School, University of Melbourne, Melbourne, VIC, Australia
| | - Kelly J. Stanger
- Australian Centre for Disease Preparedness, CSIRO, Geelong, VIC, Australia
| |
Collapse
|
12
|
Zhang M, Yin Y. Dual roles of anesthetics in postoperative cognitive dysfunction: Regulation of microglial activation through inflammatory signaling pathways. Front Immunol 2023; 14:1102312. [PMID: 36776829 PMCID: PMC9911670 DOI: 10.3389/fimmu.2023.1102312] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a prevalent clinical entity following surgery and is characterized by declined neurocognitive function. Neuroinflammation mediated by microglia is the essential mechanism of POCD. Anesthetics are thought to be a major contributor to the development of POCD, as they promote microglial activation and induce neuroinflammation. However, this claim remains controversial. Anesthetics can exert both anti- and pro-inflammatory effects by modulating microglial activation, suggesting that anesthetics may play dual roles in the pathogenesis of POCD. Here, we review the mechanisms by which the commonly used anesthetics regulate microglial activation via inflammatory signaling pathways, showing both anti- and pro-inflammatory properties of anesthetics, and indicating how perioperative administration of anesthetics might either relieve or worsen POCD development. The potential for anesthetics to enhance cognitive performance based on their anti-inflammatory properties is further discussed, emphasizing that the beneficial effects of anesthetics vary depending on dose, exposure time, and patients' characteristics. To minimize the incidence of POCD, we recommend considering these factors to select appropriate anesthetics.
Collapse
Affiliation(s)
- Mengxue Zhang
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yiqing Yin
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
13
|
Zhang Y, Zhang J, Wu J, Zhu Q, Chen C, Li Y. Implications of gut microbiota dysbiosis and fecal metabolite changes in psychologically stressed mice. Front Microbiol 2023; 14:1124454. [PMID: 37213506 PMCID: PMC10196128 DOI: 10.3389/fmicb.2023.1124454] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/18/2023] [Indexed: 05/23/2023] Open
Abstract
Introduction Psychological stress can induce affective disorders. Gut microbiota plays a vital role in emotional function regulation; however, the association between gut microbiota and psychological stress is poorly understood. We investigated effects of psychological stress on the gut microbiome and fecal metabolites and assessed the relationship between affective disorder behavior and altered fecal microbiota. Methods A psychological stress model was established in C57BL/6J mice using a communication box. Sucrose preference test, forced swim test, and open field test helped assess anxiety- and depression-like behaviors. Fecal microbiota transplantation (FMT) was conducted using fecal samples from stressed and non-stressed mice. Moreover, 16S rRNA gene sequencing and untargeted metabolomics were performed. Results After stress exposure for 14 days, a significant increase in anxiety- and depression-like behaviors was observed. FMT of "affective disorder microbiota" from psychologically stressed mice increased stress sensitivity relative to FMT of "normal microbiota" from non-stressed mice. 16S rRNA gene sequencing revealed decreased abundance of Bacteroides, Alistipes, and Lactobacillus and increased abundance of Parasutterella and Rikenellaceae_RC9_gut_group in stressed mice; furthermore, stressed mice showed differential metabolite profiles. KEGG pathway analysis indicated that differential metabolites were chiefly involved in the downregulated pathways of α-linolenic acid metabolism, taste transduction, and galactose metabolism. Alistipes and Bacteroides were mainly positively correlated and Parasutterella was mainly negatively correlated with diverse metabolites. Discussion Our findings suggest that gut microbiome dysbiosis contributes to affective disorder development in response to psychological stress.
Collapse
Affiliation(s)
- Yi Zhang
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Jing Zhang
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Jianmin Wu
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Qinwen Zhu
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Changrong Chen
- Department of Stomatology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- *Correspondence: Changrong Chen,
| | - Yanning Li
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
- Yanning Li,
| |
Collapse
|
14
|
Brogi E, Forfori F. Anesthesia and cancer recurrence: an overview. JOURNAL OF ANESTHESIA, ANALGESIA AND CRITICAL CARE (ONLINE) 2022; 2:33. [PMID: 37386584 DOI: 10.1186/s44158-022-00060-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/05/2022] [Indexed: 07/01/2023]
Abstract
Several perioperative factors are responsible for the dysregulation or suppression of the immune system with a possible impact on cancer cell growth and the development of new metastasis. These factors have the potential to directly suppress the immune system and activate hypothalamic-pituitary-adrenal axis and the sympathetic nervous system with a consequent further immunosuppressive effect.Anesthetics and analgesics used during the perioperative period may modulate the innate and adaptive immune system, inflammatory system, and angiogenesis, with a possible impact on cancer recurrence and long-term outcome. Even if the current data are controversial and contrasting, it is crucial to increase awareness about this topic among healthcare professionals for a future better and conscious choice of anesthetic techniques.In this article, we aimed to provide an overview regarding the relationship between anesthesia and cancer recurrence. We reviewed the effects of surgery, perioperative factors, and anesthetic agents on tumor cell survival and tumor recurrence.
Collapse
Affiliation(s)
- Etrusca Brogi
- Department of Anesthesia and Intensive Care, University of Pisa, Via Paradisa 2, 56124, Pisa, Italy.
| | - Francesco Forfori
- Department of Anesthesia and Intensive Care, University of Pisa, Via Paradisa 2, 56124, Pisa, Italy
| |
Collapse
|
15
|
Wei Y, Xiao L, Fan W, Zou J, Yang H, Liu B, Ye Y, Wen D, Liao L. Astrocyte Activation, but not Microglia, Is Associated with the Experimental Mouse Model of Schizophrenia Induced by Chronic Ketamine. J Mol Neurosci 2022; 72:1902-1915. [PMID: 35802289 DOI: 10.1007/s12031-022-02046-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 06/23/2022] [Indexed: 12/15/2022]
Abstract
Ketamine is a noncompetitive antagonist of N-methyl-D-aspartate (NMDA) receptors. Many experimental studies have shown that ketamine can induce cognitive impairments and schizophrenia-like symptoms. While much data have demonstrated that glial cells are associated with the pathophysiology of psychiatric disorders, including schizophrenia, the response of glial cells to ketamine and its significance to schizophrenia are not clear. The present study was intended to explore whether chronic ketamine treatment would induce behavioral and glial changes in mice. First, ketamine was used to stimulate behavioral abnormalities similar to schizophrenia evaluated by the open field test, elevated plus-maze test, Y maze test, novel object recognition test, and tail suspension test. Secondly, histopathology and Nissl staining were performed. Meanwhile, immunofluorescence was used to evaluate the expression levels of IBA-1 (a microglial marker) and GFAP (an astrocyte marker) in the mouse hippocampus for any change. Then, ELISA was used to analyze proinflammatory cytokine levels for any change. Our results showed that ketamine (25 mg/kg, i.p., qid, 12 days) induced anxiety, recognition deficits, and neuronal injury in the hippocampus. Moreover, chronic ketamine treatment enhanced GFAP expression in CA1 and DG regions of the hippocampus but did not influence the expression of IBA-1. Ketamine also increased the levels of IL-1β, IL-6, and TNF-α in the mouse hippocampus. Our study created a new procedure for ketamine administration, which successfully induce negative symptoms and cognitive-behavioral defects in schizophrenia by chronic ketamine. This study further revealed that an increase in astrocytosis, but not microglia, is associated with the mouse model of schizophrenia caused by ketamine. In summary, hippocampal astrocytes may be involved in the pathophysiology of ketamine-induced schizophrenia-like phenotypes through reactive transformation and regulation of neuroinflammation.
Collapse
Affiliation(s)
- Ying Wei
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
- College of Pharmacy, North Sichuan Medical College, Nanchong, China
| | - Li Xiao
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Weihao Fan
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Jing Zou
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Hong Yang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Bo Liu
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yi Ye
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Di Wen
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Linchuan Liao
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.
| |
Collapse
|
16
|
Zhang A, Liu Z, Liang M. A Scientometric Visualization Analysis for Molecular Mechanisms of Substance Abuse and Its Neurotoxicity From 1997 to 2021. Front Mol Neurosci 2022; 15:885701. [PMID: 35845612 PMCID: PMC9283979 DOI: 10.3389/fnmol.2022.885701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/03/2022] [Indexed: 11/13/2022] Open
Abstract
Substance abuse has become a global problem due to drug-induced addiction and neurotoxicity, which causes a huge physical, social, and financial burden. Various kinds of drugs can hijack the users’/abusers’ behavior and associated neurocircuitry. To summarize recent scientific advances on drug abuse, we reviewed relevant publications to analyze research progress and such trends through bibliometric ways. Based on retrieval strategies, a total of 681 scientific records published from 1997 to 2021 were screened and included in the Web of Science (WoS) database. Further scientometric analysis revealed that annual publication output increased across this period, with the United States of America (USA) contributing a significant number of reasons. Research has focused on neurotransmitter, oxidative stress, mitochondrial system injury, and other neurotoxic mechanisms. Neuroimmune, neurotoxic targets, and new psychoactive substances have been hot topics in recent years, which deserve continued research in the future. Specific research on molecular mechanisms has progressed across this period, with an emphasis on the root cause of toxicity and molecular targets for therapy. Moreover, collaborations of international multi-disciplinary research teams have been efficient and need to be encouraged for addiction research and the development of appropriate therapeutic processes.
Collapse
|
17
|
Białoń M, Wąsik A. Advantages and Limitations of Animal Schizophrenia Models. Int J Mol Sci 2022; 23:5968. [PMID: 35682647 PMCID: PMC9181262 DOI: 10.3390/ijms23115968] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/17/2022] [Accepted: 05/23/2022] [Indexed: 12/16/2022] Open
Abstract
Mental illness modeling is still a major challenge for scientists. Animal models of schizophrenia are essential to gain a better understanding of the disease etiopathology and mechanism of action of currently used antipsychotic drugs and help in the search for new and more effective therapies. We can distinguish among pharmacological, genetic, and neurodevelopmental models offering various neuroanatomical disorders and a different spectrum of symptoms of schizophrenia. Modeling schizophrenia is based on inducing damage or changes in the activity of relevant regions in the rodent brain (mainly the prefrontal cortex and hippocampus). Such artificially induced dysfunctions approximately correspond to the lesions found in patients with schizophrenia. However, notably, animal models of mental illness have numerous limitations and never fully reflect the disease state observed in humans.
Collapse
Affiliation(s)
| | - Agnieszka Wąsik
- Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Cracow, Poland;
| |
Collapse
|
18
|
Spencer HF, Berman RY, Boese M, Zhang M, Kim SY, Radford KD, Choi KH. Effects of an intravenous ketamine infusion on inflammatory cytokine levels in male and female Sprague-Dawley rats. J Neuroinflammation 2022; 19:75. [PMID: 35379262 PMCID: PMC8981848 DOI: 10.1186/s12974-022-02434-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 03/20/2022] [Indexed: 12/29/2022] Open
Abstract
Background Ketamine, a multimodal dissociative anesthetic drug, is widely used as an analgesic following traumatic injury. Although ketamine may produce anti-inflammatory effects when administered after injury, the immunomodulatory properties of intravenous (IV) ketamine in a non-inflammatory condition are unclear. In addition, most preclinical studies use an intraperitoneal (IP) injection of ketamine, which limits its clinical translation as patients usually receive an IV ketamine infusion after injury. Methods Here, we administered sub-anesthetic doses of a single IV ketamine infusion (0, 10, or 40 mg/kg) to male and female Sprague–Dawley rats over a 2-h period. We collected blood samples at 2- and 4-h post-ketamine infusion to determine plasma inflammatory cytokine levels using multiplex immunoassays. Results The 10 mg/kg ketamine infusion reduced spontaneous locomotor activity in male and female rats, while the 40 mg/kg infusion stimulated activity in female, but not male, rats. The IV ketamine infusion produced dose-dependent and sex-specific effects on plasma inflammatory cytokine levels. A ketamine infusion reduced KC/GRO and tumor necrosis factor alpha (TNF-α) levels in both male and female rats, interleukin-6 (IL-6) levels in female rats, and interleukin-10 (IL-10) levels in male rats. However, most cytokine levels returned to control levels at 4-h post-infusion, except for IL-6 levels in male rats and TNF-α levels in female rats, indicating a different trajectory of certain cytokine changes over time following ketamine administration. Conclusions The current findings suggest that sub-anesthetic doses of an IV ketamine infusion may produce sex-related differences in the effects on peripheral inflammatory markers in rodents, and further research is warranted to determine potential therapeutic effects of an IV ketamine infusion in an inflammatory condition.
Collapse
Affiliation(s)
- Haley F Spencer
- Program in Neuroscience, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.,Center for the Study of Traumatic Stress, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Rina Y Berman
- Center for the Study of Traumatic Stress, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Martin Boese
- Daniel K. Inouye Graduate School of Nursing, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Michael Zhang
- Center for the Study of Traumatic Stress, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Sharon Y Kim
- Program in Neuroscience, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Kennett D Radford
- Daniel K. Inouye Graduate School of Nursing, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Kwang H Choi
- Program in Neuroscience, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA. .,Center for the Study of Traumatic Stress, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA. .,Daniel K. Inouye Graduate School of Nursing, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA. .,Department of Psychiatry, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
19
|
Combination of electroconvulsive stimulation with ketamine or escitalopram protects the brain against inflammation and oxidative stress induced by maternal deprivation and is critical for associated behaviors in male and female rats. Mol Neurobiol 2022; 59:1452-1475. [PMID: 34994953 DOI: 10.1007/s12035-021-02718-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 12/23/2021] [Indexed: 12/24/2022]
Abstract
This study aimed at evaluating the treatment effects with ketamine, electroconvulsive stimulation (ECS), escitalopram, alone or in combination in adult rats of both sexes, subjected to the animal model of maternal deprivation (MD). All groups were subjected to the forced swimming test (FST), splash and open field tests. The prefrontal cortex (PFC), hippocampus and serum were collected to analyze oxidative stress and inflammatory parameters. MD induced depressive-like behavior in the FST test in males and reduced grooming time in male and female rats. The treatments alone or combined reversed depressive and anhedonic behavior in females. In males, all treatments increased grooming time, except for ECS + escitalopram + ketamine. MD increased lipid peroxidation and protein carbonylation, nitrite/nitrate concentration and myeloperoxidase activity in the PFC and hippocampus of males and females. However, the treatment's response was sex dependent. Catalase activity decreased in the PFC of males and the PFC and hippocampus of females, and most treatments were not able to reverse it. MD increased the inflammation biomarkers levels in the PFC and hippocampus of males and females, and most treatments were able to reverse this increase. In all groups, a reduction in the interleukin-10 levels in the PFC and hippocampus of female and male rats was observed. Our study shows different responses between the sexes in the patterns evaluated and reinforces the use of the gender variable as a biological factor in MDD related to early stress and in the response of the therapeutic strategies used.
Collapse
|
20
|
Kojic M, Saelens J, Kadriu B, Zarate CA, Kraus C. Ketamine for Depression: Advances in Clinical Treatment, Rapid Antidepressant Mechanisms of Action, and a Contrast with Serotonergic Psychedelics. Curr Top Behav Neurosci 2022; 56:141-167. [PMID: 35312993 PMCID: PMC10500612 DOI: 10.1007/7854_2022_313] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The approval of ketamine for treatment-resistant depression has created a model for a novel class of rapid-acting glutamatergic antidepressants. Recent research into other novel rapid-acting antidepressants - most notably serotonergic psychedelics (SPs) - has also proven promising. Presently, the mechanisms of action of these substances are under investigation to improve these novel treatments, which also exhibit considerable side effects such as dissociation. This chapter lays out the historical development of ketamine as an antidepressant, outlines its efficacy and safety profile, reviews the evidence for ketamine's molecular mechanism of action, and compares it to the proposed mechanism of SPs. The evidence suggests that although ketamine and SPs act on distinct primary targets, both may lead to rapid restoration of synaptic deficits and downstream network reconfiguration. In both classes of drugs, a glutamate surge activates α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) throughput and increases in brain-derived neurotrophic factor (BDNF) levels. Taken together, these novel antidepressant mechanisms may serve as a framework to explain the rapid and sustained antidepressant effects of ketamine and may be crucial for developing new rapid-acting antidepressants with an improved side effect profile.
Collapse
Affiliation(s)
- Marina Kojic
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Johan Saelens
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Bashkim Kadriu
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
- Department of Neuroscience, Janssen Research & Development, LLC, San Diego, CA, USA
| | - Carlos A Zarate
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Christoph Kraus
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria.
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
21
|
Choudhury D, Autry AE, Tolias KF, Krishnan V. Ketamine: Neuroprotective or Neurotoxic? Front Neurosci 2021; 15:672526. [PMID: 34566558 PMCID: PMC8461018 DOI: 10.3389/fnins.2021.672526] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 07/12/2021] [Indexed: 12/20/2022] Open
Abstract
Ketamine, a non-competitive N-methyl-D-aspartate receptor (NMDAR) antagonist, has been employed clinically as an intravenous anesthetic since the 1970s. More recently, ketamine has received attention for its rapid antidepressant effects and is actively being explored as a treatment for a wide range of neuropsychiatric syndromes. In model systems, ketamine appears to display a combination of neurotoxic and neuroprotective properties that are context dependent. At anesthetic doses applied during neurodevelopmental windows, ketamine contributes to inflammation, autophagy, apoptosis, and enhances levels of reactive oxygen species. At the same time, subanesthetic dose ketamine is a powerful activator of multiple parallel neurotrophic signaling cascades with neuroprotective actions that are not always NMDAR-dependent. Here, we summarize results from an array of preclinical studies that highlight a complex landscape of intracellular signaling pathways modulated by ketamine and juxtapose the somewhat contrasting neuroprotective and neurotoxic features of this drug.
Collapse
Affiliation(s)
- Divya Choudhury
- Department of BioSciences, Rice University, Houston, TX, United States
| | - Anita E. Autry
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Kimberley F. Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Vaishnav Krishnan
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
- Department of Neurology, Baylor College of Medicine, Houston, TX, United States
- Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
22
|
Lin Q, Fu Q, Chen D, Yu B, Luo Y, Huang Z, Zheng P, Mao X, Yu J, Luo J, Yan H, He J. Functional Characterization of Porcine NK-Lysin: A Novel Immunomodulator That Regulates Intestinal Inflammatory Response. Molecules 2021; 26:molecules26144242. [PMID: 34299517 PMCID: PMC8307250 DOI: 10.3390/molecules26144242] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/01/2021] [Accepted: 07/09/2021] [Indexed: 11/16/2022] Open
Abstract
Porcine NK-Lysine (PNKL) is a new antimicrobial peptide (AMP) identified in the small intestine. In this study, PNKL protein was obtained through heterologous expression in Escherichia coli and was estimated by SDS-PAGE at 33 kDa. The antibacterial activities of PNKL were determined using various bacterial strains and showed broad-spectrum antimicrobial activity against Gram-negative and Gram-positive bacteria. Furthermore, E. coli K88-challenged IPEC-J2 cells were used to determine PNKL influences on inflammatory responses. Hemolytic assays showed that PNKL had no detrimental impact on cell viability. Interestingly, PNKL elevated the viability of IPEC-J2 cells exposure to E. coli K88. PNKL significantly decreased the cell apoptosis rate, and improved the distribution and abundance of tight junction protein ZO-1 in IPEC-J2 cells upon E. coli K88-challenge. Importantly, PNKL not only down regulated the expressions of inflammatory cytokines such as the IL-6 and TNF-α, but also down regulated the expressions of NF-κB, Caspase3, and Caspase9 in the E. coli K88-challenged cells. These results suggest a novel function of natural killer (NK)-lysin, and the anti-bacterial and anti-inflammatory properties of PNKL may allow it a potential substitute for conventionally used antibiotics or drugs.
Collapse
Affiliation(s)
- Qian Lin
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.L.); (Q.F.); (D.C.); (B.Y.); (Y.L.); (Z.H.); (P.Z.); (X.M.); (J.Y.); (J.L.); (H.Y.)
- Key Laboratory of Animal Disease-Resistant Nutrition, Chengdu 611130, China
| | - Qingqing Fu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.L.); (Q.F.); (D.C.); (B.Y.); (Y.L.); (Z.H.); (P.Z.); (X.M.); (J.Y.); (J.L.); (H.Y.)
- Key Laboratory of Animal Disease-Resistant Nutrition, Chengdu 611130, China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.L.); (Q.F.); (D.C.); (B.Y.); (Y.L.); (Z.H.); (P.Z.); (X.M.); (J.Y.); (J.L.); (H.Y.)
- Key Laboratory of Animal Disease-Resistant Nutrition, Chengdu 611130, China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.L.); (Q.F.); (D.C.); (B.Y.); (Y.L.); (Z.H.); (P.Z.); (X.M.); (J.Y.); (J.L.); (H.Y.)
- Key Laboratory of Animal Disease-Resistant Nutrition, Chengdu 611130, China
| | - Yuheng Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.L.); (Q.F.); (D.C.); (B.Y.); (Y.L.); (Z.H.); (P.Z.); (X.M.); (J.Y.); (J.L.); (H.Y.)
- Key Laboratory of Animal Disease-Resistant Nutrition, Chengdu 611130, China
| | - Zhiqing Huang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.L.); (Q.F.); (D.C.); (B.Y.); (Y.L.); (Z.H.); (P.Z.); (X.M.); (J.Y.); (J.L.); (H.Y.)
- Key Laboratory of Animal Disease-Resistant Nutrition, Chengdu 611130, China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.L.); (Q.F.); (D.C.); (B.Y.); (Y.L.); (Z.H.); (P.Z.); (X.M.); (J.Y.); (J.L.); (H.Y.)
- Key Laboratory of Animal Disease-Resistant Nutrition, Chengdu 611130, China
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.L.); (Q.F.); (D.C.); (B.Y.); (Y.L.); (Z.H.); (P.Z.); (X.M.); (J.Y.); (J.L.); (H.Y.)
- Key Laboratory of Animal Disease-Resistant Nutrition, Chengdu 611130, China
| | - Jie Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.L.); (Q.F.); (D.C.); (B.Y.); (Y.L.); (Z.H.); (P.Z.); (X.M.); (J.Y.); (J.L.); (H.Y.)
- Key Laboratory of Animal Disease-Resistant Nutrition, Chengdu 611130, China
| | - Junqiu Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.L.); (Q.F.); (D.C.); (B.Y.); (Y.L.); (Z.H.); (P.Z.); (X.M.); (J.Y.); (J.L.); (H.Y.)
- Key Laboratory of Animal Disease-Resistant Nutrition, Chengdu 611130, China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.L.); (Q.F.); (D.C.); (B.Y.); (Y.L.); (Z.H.); (P.Z.); (X.M.); (J.Y.); (J.L.); (H.Y.)
- Key Laboratory of Animal Disease-Resistant Nutrition, Chengdu 611130, China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Q.L.); (Q.F.); (D.C.); (B.Y.); (Y.L.); (Z.H.); (P.Z.); (X.M.); (J.Y.); (J.L.); (H.Y.)
- Key Laboratory of Animal Disease-Resistant Nutrition, Chengdu 611130, China
- Correspondence: ; Tel.: +86-28-8629-1781; Fax: +86-28-8629-0922
| |
Collapse
|
23
|
Mathew AS, Gorick CM, Thim EA, Garrison WJ, Klibanov AL, Miller GW, Sheybani ND, Price RJ. Transcriptomic response of brain tissue to focused ultrasound-mediated blood-brain barrier disruption depends strongly on anesthesia. Bioeng Transl Med 2021; 6:e10198. [PMID: 34027087 PMCID: PMC8126816 DOI: 10.1002/btm2.10198] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/19/2020] [Accepted: 10/21/2020] [Indexed: 12/16/2022] Open
Abstract
Focused ultrasound (FUS) mediated blood-brain barrier disruption (BBBD) targets the delivery of systemically-administered therapeutics to the central nervous system. Preclinical investigations of BBBD have been performed on different anesthetic backgrounds; however, the influence of the choice of anesthetic on the molecular response to BBBD is unknown, despite its potential to critically affect interpretation of experimental therapeutic outcomes. Here, using bulk RNA sequencing, we comprehensively examined the transcriptomic response of both normal brain tissue and brain tissue exposed to FUS-induced BBBD in mice anesthetized with either isoflurane with medical air (Iso) or ketamine/dexmedetomidine (KD). In normal murine brain tissue, Iso alone elicited minimal differential gene expression (DGE) and repressed pathways associated with neuronal signaling. KD alone, however, led to massive DGE and enrichment of pathways associated with protein synthesis. In brain tissue exposed to BBBD (1 MHz, 0.5 Hz pulse repetition frequency, 0.4 MPa peak-negative pressure), we systematically evaluated the relative effects of anesthesia, microbubbles, and FUS on the transcriptome. Of particular interest, we observed that gene sets associated with sterile inflammatory responses and cell-cell junctional activity were induced by BBBD, regardless of the choice of anesthesia. Meanwhile, gene sets associated with metabolism, platelet activity, tissue repair, and signaling pathways, were differentially affected by BBBD, with a strong dependence on the anesthetic. We conclude that the underlying transcriptomic response to FUS-mediated BBBD may be powerfully influenced by anesthesia. These findings raise considerations for the translation of FUS-BBBD delivery approaches that impact, in particular, metabolism, tissue repair, and intracellular signaling.
Collapse
Affiliation(s)
- Alexander S. Mathew
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Catherine M. Gorick
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - E. Andrew Thim
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - William J. Garrison
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Radiology & Medical ImagingUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Alexander L. Klibanov
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Internal Medicine, Cardiovascular DivisionUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - G. Wilson Miller
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Radiology & Medical ImagingUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Natasha D. Sheybani
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Richard J. Price
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Radiology & Medical ImagingUniversity of VirginiaCharlottesvilleVirginiaUSA
| |
Collapse
|
24
|
Huang J, Xu Y, Wang F, Wang H, Li L, Deng Y, Cai L. Long Noncoding RNA SPRY4-IT1 Modulates Ketamine-Induced Neurotoxicity in Human Embryonic Stem Cell-Derived Neurons through EZH2. Dev Neurosci 2021; 43:9-17. [PMID: 33827085 DOI: 10.1159/000513535] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/02/2020] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE This study aimed to investigate whether long noncoding RNA sprouty receptor tyrosine kinase signaling antagonist 4-intronic transcript 1 (SPRY4-IT1) is involved in the regulation of ketamine-induced neurotoxicity. METHODS Human embryonic stem cells (hESCs) were induced into neurons in vitro and treated with ketamine. Apoptosis and neurite degeneration assays were used to determine ketamine-induced neurotoxicity and qRT-PCR to determine SPRY4-IT1 expression. SPRY4-IT1 was downregulated in hESC-induced neurons to examine its regulation on ketamine-induced neurotoxicity. The correlation between enhancer of zeste homolog 2 (EZH2) and SPRY4-IT1 was also examined. EZH2 was upregulated in SPRY4-IT1-downregualted hESC-induced neurons to further examine its participation in SPRY4-IT1-mediated ketamine neurotoxicity. RESULTS Ketamine-induced dose-dependent apoptosis, neurite degeneration, and SPRY4-IT1 upregulation in hESC-induced neurons. Lentivirus-mediated SPRY4-IT1 downregulation protected ketamine neurotoxicity. EZH2 expression was positively correlated with SPRY4-IT1 in hESC-induced neurons. EZH2 overexpression markedly reversed the protective effects of SPRY4-IT1 knockdown on ketamine neurotoxicity. CONCLUSIONS SPRY4-IT1 is involved in anesthesia-induced neurotoxicity, possibly through the regulation on EZH2 gene.
Collapse
Affiliation(s)
- Jingyuan Huang
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yan Xu
- Department of Anesthesiology, Xi'an Central Hospital, Xi'an, China
| | - Fang Wang
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Haili Wang
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Lu Li
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yanan Deng
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Liang Cai
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
25
|
Hernández-Avalos I, Flores-Gasca E, Mota-Rojas D, Casas-Alvarado A, Miranda-Cortés AE, Domínguez-Oliva A. Neurobiology of anesthetic-surgical stress and induced behavioral changes in dogs and cats: A review. Vet World 2021; 14:393-404. [PMID: 33776304 PMCID: PMC7994130 DOI: 10.14202/vetworld.2021.393-404] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 01/08/2021] [Indexed: 12/14/2022] Open
Abstract
The anesthetic-surgical stress response consists of metabolic, neuroendocrine, hemodynamic, immunological, and behavioral adaptations through chemical mediators such as the adrenocorticotropic hormone, growth hormone, antidiuretic hormone, cortisol, aldosterone, angiotensin II, thyroid-stimulating hormone, thyroxine, triiodothyronine, follicle-stimulating hormone, luteinizing hormone, catecholamines, insulin, interleukin (IL)-1, IL-6, tumor necrosis factor-alpha, and prostaglandin E-2. Behavioral changes include adopting the so-called prayer posture, altered facial expressions, hyporexia or anorexia, drowsiness, sleep disorders, restriction of movement, licking or biting the injured area, and vocalizations. Overall, these changes are essential mechanisms to counteract harmful stimuli. However, if uncontrolled surgical stress persists, recovery time may be prolonged, along with increased susceptibility to infections in the post-operative period. This review discusses the neurobiology and most relevant organic responses to pain and anesthetic-surgical stress in dogs and cats. It highlights the role of stress biomarkers and their influence on autonomous and demeanor aspects and emphasizes the importance of understanding and correlating all factors to provide a more accurate assessment of pain and animal welfare in dogs and cats throughout the surgical process.
Collapse
Affiliation(s)
- I Hernández-Avalos
- Department of Biological Sciences, Clinical Pharmacology and Veterinary Anesthesia, Faculty of Higher Studies Cuautitlán, Universidad Nacional Autónoma de México, State of Mexico 54714, Mexico
| | - E Flores-Gasca
- Department of Veterinary Surgery, Faculty of Higher Studies Cuautitlán, Universidad Nacional Autónoma de México, State of Mexico 54714, Mexico
| | - D Mota-Rojas
- Neurophysiology of Pain, Behavior and Assessment of Welfare in Domestic Animals, DPAA, Universidad Autónoma Metropolitana, Mexico City 04960, Mexico
| | - A Casas-Alvarado
- Master in Agricultural Sciences. Animal Welfare, Universidad Autónoma Metropolitana, Mexico City 04960, Mexico
| | - A E Miranda-Cortés
- Department of Biological Sciences, Clinical Pharmacology and Veterinary Anesthesia, Faculty of Higher Studies Cuautitlán, Universidad Nacional Autónoma de México, State of Mexico 54714, Mexico
| | - A Domínguez-Oliva
- Department of Biological Sciences, Clinical Pharmacology and Veterinary Anesthesia, Faculty of Higher Studies Cuautitlán, Universidad Nacional Autónoma de México, State of Mexico 54714, Mexico
| |
Collapse
|
26
|
Alqahtani F, Assiri MA, Mohany M, Imran I, Javaid S, Rasool MF, Shakeel W, Sivandzade F, Alanazi AZ, Al-Rejaie SS, Alshammari MA, Alasmari F, Alanazi MM, Alamri FF. Coadministration of Ketamine and Perampanel Improves Behavioral Function and Reduces Inflammation in Acute Traumatic Brain Injury Mouse Model. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3193725. [PMID: 33381547 PMCID: PMC7749776 DOI: 10.1155/2020/3193725] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/23/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) is among the most debilitating neurological disorders with inadequate therapeutic options. It affects all age groups globally leading to post-TBI behavioral challenges and life-long disabilities requiring interventions for these health issues. In the current study, C57BL/6J mice were induced with TBI through the weight-drop method, and outcomes of acutely administered ketamine alone and in combination with perampanel were observed. The impact of test drugs was evaluated for post-TBI behavioral changes by employing the open field test (OFT), Y-maze test, and novel object recognition test (NOR). After that, isolated plasma and brain homogenates were analyzed for inflammatory modulators, i.e., NF-κB and iNOS, through ELISA. Moreover, metabolomic studies were carried out to further authenticate the TBI rescuing potential of drugs. The animals treated with ketamine-perampanel combination demonstrated improved exploratory behavior in OFT (P < 0.05), while ketamine alone as well as in combination yielded anxiolytic effect (P < 0.05-0.001) in posttraumatic mice. Similarly, the % spontaneous alternation and % discrimination index were increased after the administration of ketamine alone (P < 0.05) and ketamine-perampanel combination (P < 0.01-0.001) in the Y-maze test and NOR test, respectively. ELISA demonstrated the reduced central and peripheral expression of NF-κB (P < 0.05) and iNOS (P < 0.01-0.0001) after ketamine-perampanel polypharmacy. The TBI-imparted alteration in plasma metabolites was restored by drug combination as evidenced by metabolomic studies. The outcomes were fruitful with ketamine, but the combination therapy proved more significant in improving all studied parameters. The benefits of this new investigated polypharmacy might be due to their antiglutamatergic, antioxidant, and neuroprotective capacity.
Collapse
Affiliation(s)
- Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed A. Assiri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohamed Mohany
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Imran Imran
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Sana Javaid
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
- Department of Pharmacy, The Women University, Multan 60000, Pakistan
| | - Muhammad Fawad Rasool
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Waleed Shakeel
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Farzane Sivandzade
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Ahmed Z. Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Salim S. Al-Rejaie
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Musaad A. Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed Mufadhe Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Faisal F. Alamri
- College of Sciences and Health Profession, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| |
Collapse
|
27
|
Li Y, Wen G, Ding R, Ren X, Jing C, Liu L, Yao J, Zhang G, Lu Y, Li B, Wu X. Effects of Single-Dose and Long-Term Ketamine Administration on Tau Phosphorylation-Related Enzymes GSK-3β, CDK5, PP2A, and PP2B in the Mouse Hippocampus. J Mol Neurosci 2020; 70:2068-2076. [PMID: 32705526 DOI: 10.1007/s12031-020-01613-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/22/2020] [Indexed: 10/23/2022]
Abstract
Ketamine is a recreational drug that causes emotional and cognitive impairments, but its specific mechanisms of action are still unclear. Recent evidence suggests that Tau protein phosphorylation and targeted delivery to the postsynaptic area are closely related to its neurotoxicity, and our recent studies have shown that long-term ketamine administration causes excessive Tau protein phosphorylation. However, the regulatory mechanism of Tau protein phosphorylation induced by ketamine has not been clarified. In the present study, we administered a single ketamine injection and long-term (6 months) ketamine injections in C57BL/6 mice, to investigate the effects of different doses of ketamine on the expression levels of Tau protein and its phosphorylation, the expression levels and activities of the related protein phosphokinases GSK-3β and CDK5, and the expression levels and activities of the related protein phosphatases PP2A and PP2B in the mouse hippocampus. Our results showed that both single-dose and long-term ketamine administration induced excessive phosphorylation of the Tau protein at ser202/thr205 and ser396. A single ketamine administration caused an increase in the activity of GSK-3β (at high doses) and a decrease in the activity of PP2A. On the other hand, long-term ketamine administration resulted in an increase in the activities of GSK-3β (at high doses) and CDK5, and a decrease in the activity of PP2A. Our results indicate that GSK-3β, CDK5, and PP2A may be involved in ketamine-induced Tau protein phosphorylation.
Collapse
Affiliation(s)
- Yanning Li
- School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China
- School of Basic Medicine, Gannan Medical University, Ganzhou, People's Republic of China
| | - Gehua Wen
- School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China
| | - Runtao Ding
- School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China
| | - Xinghua Ren
- School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China
| | - Chenchen Jing
- School of Basic Medicine, Gannan Medical University, Ganzhou, People's Republic of China
| | - Lin Liu
- School of Basic Medicine, Gannan Medical University, Ganzhou, People's Republic of China
| | - Jun Yao
- School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China
| | - Guohua Zhang
- School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China
| | - Yan Lu
- Key Laboratory of Health Ministry in Congenital Malformation, The Affiliated Shengjing Hospital of China Medical University, Shenyang, People's Republic of China.
| | - Baoman Li
- School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China.
| | - Xu Wu
- School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China.
| |
Collapse
|
28
|
Lisowska B, Jakubiak J, Siewruk K, Sady M, Kosson D. Which idea is better with regard to immune response? Opioid anesthesia or opioid free anesthesia. J Inflamm Res 2020; 13:859-869. [PMID: 33177861 PMCID: PMC7652233 DOI: 10.2147/jir.s275986] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022] Open
Abstract
The stress of surgery is characterized by an inflammatory response with immune suppression resulting from many factors, including the type of surgery and the kind of anesthesia, linked with the drugs that are used and the underlying disease of the patient. The trauma of surgery triggers a cascade of reactions involving the immune response and nociception. As strong analgesics, opioids provide the analgesic component of general anesthesia with bi-directional effect on the immune system. Opioids influence almost all aspects of the immune response in regards to leukocytes, macrophages, mast cells, lymphocytes, and NK cells. The suppressive effect of opioids on the immune system is limiting their use, especially in patients with impaired immune response, so the possibility of using multimodal anesthesia without opioids, known as opioid-free anesthesia (OFA), is gaining more and more sympathizers. The idea of OFA is to eliminate opioid analgesia in the treatment of acute pain and to replace it with drugs from other groups that are assumed to have a comparable analgesic effect without affecting the immune system. Here, we present a review on the impact of anesthesia, with and without the use of opioids, on the immune response to surgical stress.
Collapse
Affiliation(s)
- Barbara Lisowska
- Department Anesthesiology and Intensive Medical Care, National Geriatrics, Rheumatology and Rehabilitation Institute, Warsaw 02-637, Poland
| | - Jakub Jakubiak
- Department of Anesthesiology and Intensive Care, John Paul II Western Hospital, Grodzisk Mazowiecki 05-825, Poland
| | - Katarzyna Siewruk
- Faculty of Veterinary Medicine, Department of Large Animal Diseases with Clinic, Warsaw University of Life Sciences, Warsaw 02-797, Poland
| | - Maria Sady
- Faculty of Veterinary Medicine, Department of Large Animal Diseases with Clinic, Warsaw University of Life Sciences, Warsaw 02-797, Poland
| | - Dariusz Kosson
- Department of Anaesthesiology and Intensive Care, Division of Teaching, Medical University of Warsaw, Warsaw 02-005, Poland
| |
Collapse
|
29
|
Abstract
An intranasal formulation of esketamine, the S enantiomer of ketamine, in conjunction with an oral antidepressant, has been approved by the FDA for treating treatment-resistant major depressive disorder (TRD) in 2019, almost 50 years after it was approved as an intravenous anesthetic. In contrast to traditional antidepressants, ketamine shows a rapid (within 2 h) and sustained (∼7 days) antidepressant effect and has significant positive effects on antisuicidal ideation. Ketamine's antidepressant mechanism is predominantly mediated by the N-methyl-d-aspartate receptor (NMDA) receptor, although NMDA-independent mechanisms are not ruled out. At the neurocircuitry level, ketamine affects the brain's reward and mood circuitry located in the corticomesolimbic structures involving the hippocampus, nucleus accumbens, and prefrontal cortex. Repurposing of ketamine for treating TRD provided a new understanding of the pathophysiology of depression, a paradigm shift from monoamine to glutamatergic neurotransmission, thus making it a unique tool to investigate the brain and its complex neurocircuitries.
Collapse
Affiliation(s)
- Joydip Das
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas 77204, United States
| |
Collapse
|
30
|
Binge and Subchronic Exposure to Ketamine Promote Memory Impairments and Damages in the Hippocampus and Peripheral Tissues in Rats: Gallic Acid Protective Effects. Neurotox Res 2020; 38:274-286. [DOI: 10.1007/s12640-020-00215-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/14/2020] [Accepted: 04/22/2020] [Indexed: 12/11/2022]
|
31
|
Wu Z, Zhang Y, Zhang Y, Zhao P. Sirtuin 2 Inhibition Attenuates Sevoflurane-Induced Learning and Memory Deficits in Developing Rats via Modulating Microglial Activation. Cell Mol Neurobiol 2020; 40:437-446. [PMID: 31713761 PMCID: PMC11449016 DOI: 10.1007/s10571-019-00746-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/15/2019] [Indexed: 12/27/2022]
Abstract
Sevoflurane is a widely used inhalational anesthetic in pediatric medicine that has been reported to have deleterious effects on the developing brain. Strategies to mitigate these detrimental effects are lacking. Sirtuin 2 (SIRT2) is a member of nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylases involved in a wide range of pathophysiological processes. SIRT2 inhibition has emerged as a promising treatment for an array of neurological disorders. However, the direct effects of SIRT2 on anesthesia-induced damage to the immature brain are unclear. Neonatal rats were exposed to 3% sevoflurane or 30% oxygen for 2 h daily with or without SIRT2 inhibitor AK7 pretreatment from postnatal day 7 (P7) to P9. One cohort of rats were euthanized 6, 12, and/or 24 h after the last gas exposure, and brain tissues were harvested for biochemical analysis and/or immunohistochemical examination. Cognitive functions were evaluated using the open field and Morris water maze tests on P25 and P28-32, respectively. SIRT2 was significantly up-regulated in neonatal rat hippocampus at 6 and 12 h post-anesthesia. Pretreatment with SIRT2 inhibitor AK7 reversed sevoflurane-induced hippocampus-dependent cognitive impairments. Furthermore, AK7 administration mitigated sevoflurane-induced neuroinflammation and microglial activation. Concomitantly, AK7 inhibited pro-inflammatory/M1-related markers and increased anti-inflammatory/M2-related markers in microglia. AK7 might prevent sevoflurane-induced neuroinflammation by switching microglia from the M1 to M2 phenotype. Downregulation of SIRT2 may be a novel therapeutic target for alleviating anesthesia-induced developmental neurotoxicity.
Collapse
Affiliation(s)
- Ziyi Wu
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Yi Zhang
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Yinong Zhang
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Ping Zhao
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
32
|
The Acute and Early Effects of Whole-Brain Irradiation on Glial Activation, Brain Metabolism, and Behavior: a Positron Emission Tomography Study. Mol Imaging Biol 2020; 22:1012-1020. [PMID: 32052277 PMCID: PMC7343765 DOI: 10.1007/s11307-020-01483-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Purpose Radiotherapy is a frequently applied treatment modality for brain tumors. Concomitant irradiation of normal brain tissue can induce various physiological responses. The aim of this study was to investigate whether acute and early-delayed effects of brain irradiation on glial activation and brain metabolism can be detected with positron emission tomography (PET) and whether these effects are correlated with behavioral changes. Procedures Rats underwent 0-, 10-, or 25-Gy whole-brain irradiation. At 3 and 31 days post irradiation, 1-(2-chlorophenyl)-N-[11C]methyl-(1-methylpropyl)-3-isoquinoline carboxamide ([11C]PK11195) and 2-deoxy-2-[18F]fluoro-d-glucose ([18F]FDG) PET scans were acquired to detect changes in glial activation (neuroinflammation) and glucose metabolism, respectively. The open-field test (OFT) was performed on days 6 and 27 to assess behavioral changes. Results Twenty-five-gray-irradiated rats showed higher [11C]PK11195 uptake in most brain regions than controls on day 3 (striatum, hypothalamus, accumbens, septum p < 0.05), although some brain regions had lower uptake (cerebellum, parietal association/retrosplenial visual cortex, frontal association/motor cortex, somatosensory cortex, p < 0.05). On day 31, several brain regions in 25-Gy-irradiated rats still showed significantly higher [11C]PK11195 uptake than controls and 10-Gy-irradiated group (p < 0.05). Within-group analysis showed that [11C]PK11195 uptake in individual brain regions of 25-Gy treated rats remained stable or slightly increased between days 3 and 31. In contrast, a significant reduction (p < 0.05) in tracer uptake between days 3 and 31 was found in all brain areas of controls and 10-Gy-irradiated animals. Moreover, 10-Gy treatment led to a significantly higher [18F]FDG uptake on day 3 (p < 0.05). [18F]FDG uptake decreased between days 3 and 31 in all groups; no significant differences between groups were observed anymore on day 31, except for increased uptake in the hypothalamus in the 10-Gy group. The OFT did not show any significant differences between groups. Conclusions Non-invasive PET imaging indicated that brain irradiation induces neuroinflammation and a metabolic flare, without causing acute or early-delayed behavioral changes. Electronic supplementary material The online version of this article (10.1007/s11307-020-01483-y) contains supplementary material, which is available to authorized users.
Collapse
|
33
|
Gautam CS, Mahajan SS, Sharma J, Singh H, Singh J. Repurposing Potential of Ketamine: Opportunities and Challenges. Indian J Psychol Med 2020; 42:22-29. [PMID: 31997862 PMCID: PMC6970308 DOI: 10.4103/ijpsym.ijpsym_228_19] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/13/2019] [Accepted: 07/15/2019] [Indexed: 02/06/2023] Open
Abstract
Ketamine is a noncompetitive antagonist of the N-methyl-D-aspartate (NMDA) receptor which also interacts with various other receptors that account for its myriad actions. Originally approved as a general anesthetic, it is being explored to be repurposed for numerous other indications such as depressive disorders, suicidal ideation, substance-use disorders, anxiety disorders, chronic pain, refractory status epilepticus, and bronchial asthma exacerbations. Numerous trials are ongoing for the same. The nasal spray of esketamine, a more potent S (+) enantiomer of ketamine, has been approved by the United States Food and Drug Administration (USFDA) for treatment-resistant depression along with the oral antidepressants. However, there are concerns about its safety on long term use, given its psychedelic effects and potential abuse. In this review, we discuss repurposing ketamine for potential therapeutic use and about the safety concerns related to ketamine and esketamine.
Collapse
Affiliation(s)
- C. S. Gautam
- Department of Pharmacology, Government Medical College and Hospital, Chandigarh, Punjab, India
| | - Sonia S. Mahajan
- Department of Pharmacology, Government Medical College and Hospital, Chandigarh, Punjab, India
| | - Jatin Sharma
- Department of Pharmacology, Government Medical College and Hospital, Chandigarh, Punjab, India
| | - Harmanjit Singh
- Department of Pharmacology, Government Medical College and Hospital, Chandigarh, Punjab, India
| | - Jagjit Singh
- Department of Pharmacology, Government Medical College and Hospital, Chandigarh, Punjab, India
| |
Collapse
|
34
|
Schiavone S, Tucci P, Trabace L, Morgese MG. Early Celastrol Administration Prevents Ketamine-Induced Psychotic-Like Behavioral Dysfunctions, Oxidative Stress and IL-10 Reduction in The Cerebellum of Adult Mice. Molecules 2019; 24:molecules24213993. [PMID: 31694174 PMCID: PMC6864687 DOI: 10.3390/molecules24213993] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/22/2019] [Accepted: 10/25/2019] [Indexed: 12/11/2022] Open
Abstract
Administration of subanesthetic doses of ketamine during brain maturation represents a tool to mimic an early insult to the central nervous system (CNS). The cerebellum is a key player in psychosis pathogenesis, to which oxidative stress also contributes. Here, we investigated the impact of early celastrol administration on behavioral dysfunctions in adult mice that had received ketamine (30 mg/kg i.p.) at postnatal days (PNDs) 7, 9, and 11. Cerebellar levels of 8-hydroxydeoxyguanosine (8-OHdG), NADPH oxidase (NOX) 1 and NOX2, as well as of the calcium-binding protein parvalbumin (PV), were also assessed. Furthermore, celastrol effects on ketamine-induced alterations of proinflammatory (TNF-α, IL-6 and IL-1β) and anti-inflammatory (IL-10) cytokines in this brain region were evaluated. Early celastrol administration prevented ketamine-induced discrimination index decrease at adulthood. The same was found for locomotor activity elevations and increased close following and allogrooming, whereas no beneficial effects on sniffing impairment were detected. Ketamine increased 8-OHdG in the cerebellum of adult mice, which was also prevented by early celastrol injection. Cerebellar NOX1 levels were enhanced at adulthood following postnatal ketamine exposure. Celastrol per se induced NOX1 decrease in the cerebellum. This effect was more significant in animals that were early administered with ketamine. NOX2 levels did not change. Ketamine administration did not affect PV amount in the cerebellum. TNF-α levels were enhanced in ketamine-treated animals; however, this was not prevented by early celastrol administration. While no changes were observed for IL-6 and IL-1β levels, ketamine determined a reduction of cerebellar IL-10 expression, which was prevented by early celastrol treatment. Our results suggest that NOX inhibition during brain maturation prevents the development of psychotic-like behavioral dysfunctions, as well as the increased cerebellar oxidative stress and the reduction of IL-10 in the same brain region following ketamine exposure in postnatal life. This opens novel neuroprotective opportunities against early detrimental insults occurring during brain development.
Collapse
|
35
|
Frinchi M, Nuzzo D, Scaduto P, Di Carlo M, Massenti MF, Belluardo N, Mudò G. Anti-inflammatory and antioxidant effects of muscarinic acetylcholine receptor (mAChR) activation in the rat hippocampus. Sci Rep 2019; 9:14233. [PMID: 31578381 PMCID: PMC6775129 DOI: 10.1038/s41598-019-50708-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 07/19/2019] [Indexed: 12/17/2022] Open
Abstract
Recently we found that acute treatment with Oxotremorine (Oxo), a non-selective mAChRs agonist, up-regulates heat shock proteins and activates their transcription factor heat shock factor 1 in the rat hippocampus. Here we aimed to investigate: a) if acute treatment with Oxo may regulate pro-inflammatory or anti-inflammatory cytokines and oxidative stress in the rat hippocampus; b) if chronic restraint stress (CRS) induces inflammatory or oxidative alterations in the hippocampus and whether such alterations may be affected by chronic treatment with Oxo. In the acute experiment, rats were injected with single dose of Oxo (0.4 mg/kg) and sacrificed at 24 h, 48 h and 72 h. In the CRS experiment, the rats were exposed for 21 days to the CRS and then were treated with Oxo (0.2 mg/kg) for further 10 days. The acute Oxo treatment showed an ability to significantly reduce reactive oxygen species (ROS), singlet oxygen (1O2), pro-inflammatory cytokines levels (IL-1β and IL-6) and phosphorylated NF-κB-p65. Acute Oxo treatment also increased superoxide dismutase (SOD)-2 protein levels and stimulated SOD activity. No differences were detected in the anti-inflammatory cytokine levels, including IL-10 and TGF-β1. In the group of rats exposed to the CRS were found increased hippocampal IL-1β and IL-6 levels, together with a reduction of SOD activity level. These changes produced by CRS were counteracted by chronic Oxo treatment. In contrast, the upregulation of ROS and 1O2 levels in the CRS group was not counteracted by chronic Oxo treatment. The results revealed a hippocampal anti-inflammatory and antioxidant effect of Oxo treatment in both basal conditions and anti-inflammatory in the CRS rat model.
Collapse
Affiliation(s)
- Monica Frinchi
- Department of Biomedicine, Neurosciences and Advanced Diagnostic, div. of Human Physiology, University of Palermo, 90134, Palermo, Italy
| | - Domenico Nuzzo
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy" (IBIM), Consiglio Nazionale delle Ricerche (CNR), 90146, Palermo, Italy
| | - Pietro Scaduto
- Department of Biomedicine, Neurosciences and Advanced Diagnostic, div. of Human Physiology, University of Palermo, 90134, Palermo, Italy
| | - Marta Di Carlo
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy" (IBIM), Consiglio Nazionale delle Ricerche (CNR), 90146, Palermo, Italy
| | - Maria F Massenti
- Department of Sciences for Health Promotion and Mother and Child Care "Giuseppe D'Alessandro", University of Palermo, 90134, Palermo, Italy
| | - Natale Belluardo
- Department of Biomedicine, Neurosciences and Advanced Diagnostic, div. of Human Physiology, University of Palermo, 90134, Palermo, Italy
| | - Giuseppa Mudò
- Department of Biomedicine, Neurosciences and Advanced Diagnostic, div. of Human Physiology, University of Palermo, 90134, Palermo, Italy.
| |
Collapse
|
36
|
Pillinger T, Osimo EF, Brugger S, Mondelli V, McCutcheon RA, Howes OD. A Meta-analysis of Immune Parameters, Variability, and Assessment of Modal Distribution in Psychosis and Test of the Immune Subgroup Hypothesis. Schizophr Bull 2019; 45:1120-1133. [PMID: 30407606 PMCID: PMC6737479 DOI: 10.1093/schbul/sby160] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Immune parameters are elevated in psychosis, but it is unclear whether alterations are homogenous across patients or heterogeneity exists, consistent with the hypothesis that immune alterations are specific to a subgroup of patients. To address this, we examine whether antipsychotic-naïve first-episode psychosis patients exhibit greater variability in blood cytokines, C-reactive protein, and white cell counts compared with controls, and if group mean differences persist after adjusting for skewed data and potential confounds. Databases were searched for studies reporting levels of peripheral immune parameters. Means and variances were extracted and analyzed using multivariate meta-analysis of mean and variability of differences. Outcomes were (1) variability in patients relative to controls, indexed by variability ratio (VR) and coefficient of variation ratio (CVR); (2) mean differences indexed by Hedges g; (3) Modal distribution of raw immune parameter data using Hartigan's unimodality dip test. Thirty-five studies reporting on 1263 patients and 1470 controls were included. Variability of interleukin-6 (IL6) (VR = 0.19), tumor necrosis factor-α (TNFα) (VR = 0.36), interleukin-1β (VR = 0.35), interleukin-4 (VR = 0.55), and interleukin-8 (VR = 0.28) was reduced in patients. Results persisted for IL6 and IL8 after mean-scaling. Ninety-four percent and one hundred percent of raw data were unimodally distributed in psychosis and controls, respectively. Mean levels of IL6 (g = 0.62), TNFα (g = 0.56), interferon-γ (IFNγ) (g = 0.32), transforming growth factor-β (g = 0.53), and interleukin-17 (IL17) (g = 0.48) were elevated in psychosis. Sensitivity analyses indicated this is unlikely explained by confounders for IL6, IFNγ, and IL17. These findings show elevated cytokines in psychosis after accounting for confounds, and that the hypothesis of an immune subgroup is not supported by the variability or modal distribution.
Collapse
Affiliation(s)
- Toby Pillinger
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Emanuele F Osimo
- Department of Psychiatry, University of Cambridge, Cambridge, UK,Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK,Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Stefan Brugger
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK,Medical Research Council London Institute of Medical Sciences, London, UK,Division of Psychiatry, University College London, London, UK
| | - Valeria Mondelli
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK,National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King’s College London, London, UK
| | - Robert A McCutcheon
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK,Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK,Medical Research Council London Institute of Medical Sciences, London, UK
| | - Oliver D Howes
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK,Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK,Medical Research Council London Institute of Medical Sciences, London, UK,To whom correspondence should be addressed; tel: +44-207-848-0355, e-mail:
| |
Collapse
|
37
|
Sesamin Enhances Nrf2-Mediated Protective Defense against Oxidative Stress and Inflammation in Colitis via AKT and ERK Activation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2432416. [PMID: 31534619 PMCID: PMC6732632 DOI: 10.1155/2019/2432416] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/18/2019] [Accepted: 07/14/2019] [Indexed: 12/13/2022]
Abstract
Ulcerative colitis (UC) is a major form of inflammatory bowel disease (IBD) with high incidence and prevalence in many countries. Patients with UC usually suffer from a lifetime of debilitating physical symptoms. Therefore, developing effective therapeutic strategy that can manage this disease better and improve patients' life quality is in urgent need. Sesamin (SSM) is a lignan derived from sesame seeds. In this study, the protective effect of SSM against UC and the underlying mechanism were investigated in vitro and in vivo. Our data showed that SSM protected Caco-2 cells from H2O2-induced oxidative stress injury via GSH-mediated scavenging of reactive oxygen species (ROS). Dual luciferase reporter assay showed that the transcriptional activity of nuclear factor erythroid-related factor 2 (Nrf2) was significantly increased by SSM, and the ability of SSM to activate Nrf2-targeted genes was further confirmed in Caco-2 cells using western blot and quantitative real-time PCR (qRT-PCR). In contrast, Nrf2 knockdown abolished the protective effect of SSM. Additionally, we found that SSM also activated advanced protein kinase B (AKT) and extracellular signal-regulated kinase (ERK) in Caco-2 cells, while either AKT or ERK inhibition can prevent SSM-mediated nuclear translocation of Nrf2. Furthermore, SSM displayed a better protective effect against dextran sulfate sodium- (DSS-) induced UC compared with 5-aminosalicylic acid (5-ASA) in C57BL/6 mice. The enhanced Nrf2 signaling and activated AKT/ERK were also observed in the colon of mice after SSM administration. These results first demonstrate the protective effect of SSM against UC and indicate that the effect is associated with AKT/ERK activation and subsequent Nrf2 signaling enhancement. This study provides a new insight into the medicinal value of SSM and proposes it as a new natural nutrition for better managing the symptoms of UC.
Collapse
|
38
|
Li C, Peng J, Hu R, Yan J, Sun Y, Zhang L, Liu W, Jiang H. Safety and Efficacy of Ketamine Versus Ketamine-Fentanyl-Dexmedetomidine Combination for Anesthesia and Analgesia in Rats. Dose Response 2019; 17:1559325819825902. [PMID: 30792614 PMCID: PMC6376518 DOI: 10.1177/1559325819825902] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/11/2018] [Accepted: 01/02/2019] [Indexed: 12/16/2022] Open
Abstract
Ketamine (KET), a noncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist, is most frequently used as an anesthetic, analgesic, and sedative drug in pediatric clinical practices. However, the adverse effects of KET administration such as psychotic episodes limited the use of KET. The aim of the present study was to evaluate whether the addition of small doses of fentanyl (FENT) and dexmedetomidine would reduce the overall KET consumption without concession on the safety and efficacy of anesthesia and analgesia in rats. We compared the effects of KET (50 mg/kg) administration alone and KET (25 mg/kg) combined with FENT (0.005 mg/kg) and dexmedetomidine (0.05 mg/kg) (KFD) on the times of onset and duration of anesthesia and analgesia. Compared with the KET group, the KFD group provides similar onset time of anesthesia, but longer duration of anesthesia, and better analgesic effect. Unlike the KET group, the KFD group had a lower heart rate and higher respiratory rate. Meanwhile, KFD induced markedly changes in the electroencephalography (EEG) spectral power when compared with control and KET. Furthermore, combination of FENT and dexmedetomidine alleviated the liver toxicity of KET. These results indicated that, when compared with KET alone, the administration of KFD combination offered safer and more efficient anesthesia.
Collapse
Affiliation(s)
- Chunzhu Li
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiali Peng
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Hu
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia Yan
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Sun
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Zhang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenbin Liu
- Shanghai Key Laboratory of Crime Scene Evidence, Shanghai Research Institute of Criminal Science and Technology, Shanghai, China
| | - Hong Jiang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
39
|
Wen G, Yao H, Li Y, Ding R, Ren X, Tan Y, Ren W, Yu H, Zhan X, Wang X, Xu E, Yao J, Zhang G, Lu Y, Wu X. Regulation of Tau Protein on the Antidepressant Effects of Ketamine in the Chronic Unpredictable Mild Stress Model. Front Psychiatry 2019; 10:287. [PMID: 31114516 PMCID: PMC6503093 DOI: 10.3389/fpsyt.2019.00287] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 04/12/2019] [Indexed: 12/31/2022] Open
Abstract
Tau protein is known to play an important role in maintaining microtubule assembly and stabilization, and maintaining the normal morphology of neurons, but several studies have found that chronic stress leads to Tau hyperphosphorylation. A large number of clinical trials have found that ketamine, which is an N-methyl-D-aspartate receptor antagonist, produces a rapid, long-lasting, and potent antidepressant effect in patients suffering from major depression. This rapid antidepressant effect of ketamine, which involves many mechanisms, has attracted wide attention. However, the relationship between ketamine's antidepressant effects and Tau protein has rarely been examined. We used C57BL/6 and Tau KO mice exposed to 42 days of chronic unpredictable mild stress (the CUMS model) to investigate the effect of ketamine on behavioral changes and synaptic functioning of the hippocampus. The results showed that a single treatment of ketamine rapidly relieved the CUMS-induced anhedonia, depression-like, and anxious behaviors of the C57BL/6 mice. The abnormal behaviors were accompanied by increased levels of specific alterations of hyperphosphorylated Tau protein in cytoplasm and synapse in the hippocampus of the C57BL/6 mice, but ketamine reduced the aggregation of hyperphosphorylated Tau protein only in the synapse. We also found that CUMS exposure reduced the levels of GluA1 and PSD95 in the hippocampus of the C57BL/6 mice and that these deficits were reversed by ketamine. However, the Tau KO mice did not develop any stress-induced depressive behaviors or deficits of hippocampal function. The antidepressant effect of ketamine may decrease the levels of hyperphosphorylated Tau protein in synapse of C57BL/6 mice.
Collapse
Affiliation(s)
- Gehua Wen
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Hui Yao
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Yanning Li
- Department of Forensic Medicine, School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Runtao Ding
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Xinghua Ren
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Yaqing Tan
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Weishu Ren
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Hao Yu
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Xiaoni Zhan
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Xiaolong Wang
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Enyu Xu
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Jun Yao
- 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 Sheng Jing Hospital of China Medical University, Shenyang, China
| | - Xu Wu
- School of Forensic Medicine, China Medical University, Shenyang, China
| |
Collapse
|
40
|
Rainville JR, Hodes GE. Inflaming sex differences in mood disorders. Neuropsychopharmacology 2019; 44:184-199. [PMID: 29955150 PMCID: PMC6235877 DOI: 10.1038/s41386-018-0124-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 06/07/2018] [Accepted: 06/11/2018] [Indexed: 02/06/2023]
Abstract
Men and women often experience different symptoms or rates of occurrence for a variety of mood disorders. Many of the symptoms of mood disorders overlap with autoimmune disorders, which also have a higher prevalence in women. There is a growing interest in exploring the immune system to provide biomarkers for diagnosis of mood disorders, along with new targets for developing treatments. This review examines known sex differences in the immune system and their relationship to mood disorders. We focus on immune alterations associated with unipolar depression, bipolar depression, and anxiety disorders. We describe work from both basic and clinical research examining potential immune mechanisms thought to contribute to stress susceptibility and associated mood disorders. We propose that sex and age are important, intertwined factors that need to be included in future experimental designs if we are going to harness the power of the immune system to develop a new wave of treatments for mood disorders.
Collapse
Affiliation(s)
- Jennifer R Rainville
- Department of Neuroscience, Virginia Polytechnic Institute and State University, 1981 Kraft Drive, Blacksburg, VA, 24060, USA
| | - Georgia E Hodes
- Department of Neuroscience, Virginia Polytechnic Institute and State University, 1981 Kraft Drive, Blacksburg, VA, 24060, USA.
| |
Collapse
|
41
|
GSK3β: a plausible mechanism of cognitive and hippocampal changes induced by erythropoietin treatment in mood disorders? Transl Psychiatry 2018; 8:216. [PMID: 30310078 PMCID: PMC6181907 DOI: 10.1038/s41398-018-0270-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 06/11/2018] [Accepted: 07/14/2018] [Indexed: 12/16/2022] Open
Abstract
Mood disorders are associated with significant psychosocial and occupational disability. It is estimated that major depressive disorder (MDD) will become the second leading cause of disability worldwide by 2020. Existing pharmacological and psychological treatments are limited for targeting cognitive dysfunctions in mood disorders. However, growing evidence from human and animal studies has shown that treatment with erythropoietin (EPO) can improve cognitive function. A recent study involving EPO-treated patients with mood disorders showed that the neural basis for their cognitive improvements appeared to involve an increase in hippocampal volume. Molecular mechanisms underlying hippocampal changes have been proposed, including the activation of anti-apoptotic, antioxidant, pro-survival and anti-inflammatory signalling pathways. The aim of this review is to describe the potential importance of glycogen synthase kinase 3-beta (GSK3β) as a multi-potent molecular mechanism of EPO-induced hippocampal volume change in mood disorder patients. We first examine published associations between EPO administration, mood disorders, cognition and hippocampal volume. We then highlight evidence suggesting that GSK3β influences hippocampal volume in MDD patients, and how this could assist with targeting more precise treatments particularly for cognitive deficits in patients with mood disorders. We conclude by suggesting how this developing area of research can be further advanced, such as using pharmacogenetic studies of EPO treatment in patients with mood disorders.
Collapse
|
42
|
Ahmed HI, Abdel-Sattar SA, Zaky HS. Vinpocetine halts ketamine-induced schizophrenia-like deficits in rats: impact on BDNF and GSK-3β/β-catenin pathway. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:1327-1338. [PMID: 30083945 DOI: 10.1007/s00210-018-1552-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/31/2018] [Indexed: 01/03/2023]
Abstract
There are increasing evidences supporting the involvement of oxidative stress and neuroinflammation in schizophrenia. Vinpocetine, a nootropic phosphodiesterase-1 inhibitor, was proven to possess anti-oxidant and anti-inflammatory potentials. This research aimed to reveal the likely protective features of vinpocetine against ketamine-induced schizophrenia-like deficits in rats. Additionally, the probable mechanisms contributing to this neuroprotection were also elucidated. Vinpocetine was given (20 mg/kg, i.p.) once a day for 14 days commencing 7 days before administrating ketamine (25 mg/kg i.p.). Risperidone was applied as a reference antipsychotic. Vinpocetine pre-treatment revealed a marked amendment in the hyperlocomotion, anxiety, and short-term memory deficits induced by ketamine in rats. In rats' hippocampus, ketamine induced a drastic increase in tissue levels of dopamine, lipid peroxidation, and pro-inflammatory cytokines along with a significant decrease in glutamate, GABA, SOD, and total anti-oxidant capacity. Also, ketamine induced a reduced level of BDNF together with the potentiation of GSK-3β/β-catenin pathway that led to the destruction of β-catenin. Pre-treatment of ketamine-challenged animals with vinpocetine significantly attenuated oxidative stress, inflammation, and neurotransmitter alterations. Vinpocetine also elevated BDNF expression and prevented ketamine-induced stimulation of the GSK-3β/β-catenin signaling. This research presents enlightenments into the role of vinpocetine in schizophrenia. This role may be accomplished through its effect on oxidative stress, inflammation as well as modulating BDNF and the GSK-3β/β-catenin pathway.
Collapse
Affiliation(s)
- Hebatalla I Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11754, Egypt.
| | - Somaia A Abdel-Sattar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11754, Egypt
| | - Heba S Zaky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11754, Egypt
| |
Collapse
|
43
|
Sattar Y, Wilson J, Khan AM, Adnan M, Azzopardi Larios D, Shrestha S, Rahman Q, Mansuri Z, Hassan A, Patel NB, Tariq N, Latchana S, Lopez Pantoja SC, Vargas S, Shaikh NA, Syed F, Mittal D, Rumesa F. A Review of the Mechanism of Antagonism of N-methyl-D-aspartate Receptor by Ketamine in Treatment-resistant Depression. Cureus 2018; 10:e2652. [PMID: 30034974 PMCID: PMC6051558 DOI: 10.7759/cureus.2652] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 05/18/2018] [Indexed: 12/19/2022] Open
Abstract
The biochemical processes involved in depression go beyond serotonin, norepinephrine, and dopamine. The N-methyl-D-aspartate (NMDA) receptor has a major role in the neurophysiology of depression. Ketamine, one of the prototypical NMDA antagonists, works rapidly in controlling depressive symptoms, including acutely suicidal behavior, by just a single injection. Ketamine may rapidly increase the glutamate levels and lead to structural neuronal changes. Increased neuronal dendritic growth may contribute to synaptogenesis and an increase in brain-derived neurotrophic factor (BDNF). Activation of the mechanistic target of rapamycin (mTOR), as well as increased levels of BDNF, may increase long-term potentiation and result in an improvement in the symptoms of depression. The mechanisms of ketamine's proposed effect as an off-label treatment for resistant depression are outlined in this paper.
Collapse
Affiliation(s)
- Yasar Sattar
- Research Assistant, Kings County Hospital Center, New York, USA
| | - John Wilson
- Adult Psychiatry, SUNY Downstate Medical Center
| | - Ali M Khan
- Psychiatry Resident, University of Texas Rio Grande Valley, Harlingen, Texas, USA
| | - Mahwish Adnan
- Center for Addiction and Mental Health, University of Toronto, toronto, CAN
| | | | | | | | - Zeeshan Mansuri
- Psychiatry, Texas Tech University Health Sciences Center at Odessa/permian Basin
| | - Ali Hassan
- Medical Graduate, American University of Antigua
| | | | | | | | | | - Sadiasept Vargas
- Department of Medicine, Instituto Tecnológico De Santo Domingo, Santo Domingo, DOM
| | | | - Fawaduzzaman Syed
- Internal Medicine, Sindh Medical College, Dow University of Health Sciences, Chicago, USA
| | - Daaman Mittal
- Pediatrics, Punjab Institute of Medical Sciences, ludhiana, IND
| | | |
Collapse
|
44
|
Ding Y, Ren J, Yu H, Yu W, Zhou Y. Porphyromonas gingivalis, a periodontitis causing bacterium, induces memory impairment and age-dependent neuroinflammation in mice. IMMUNITY & AGEING 2018; 15:6. [PMID: 29422938 PMCID: PMC5791180 DOI: 10.1186/s12979-017-0110-7] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 12/28/2017] [Indexed: 12/16/2022]
Abstract
Background A possible relationship between periodontitis and Alzheimer's disease (AD) has been reported. However, there is limited information on the association between the Porphyromonas gingivalis (P. gingivalis) periodontal infection and the pathological features of AD. The hypothesis that P. gingivalis periodontal infection may cause cognitive impairment via age-dependent neuroinflammation was tested. Results Thirty 4-week-old (young) female C57BL/6 J mice were randomly divided into two groups, the control group and the experimental group. Thirty 12-month-old (middle-aged) were grouped as above. The mouth of the mice in the experimental group was infected with P. gingivalis. Morris water maze(MWM) was performed to assess the learning and memory ability of mice after 6 weeks. Moreover, the expression levels of the pro-inflammatory cytokines TNF-α, IL-6, and IL-1β in the mice brain tissues were determined by Quantitative real-time polymerase chain reaction (qRT-PCR), Enzyme Linked Immunosorbent Assay(ELISA) and immunohistochemistry. Our results showed that the learning and memory abilities of the middle-aged P. gingivalis infected mice were impaired. Moreover, the expression levels of the pro-inflammatory cytokines TNF-α, IL-6, and IL-1β in the brain tissues of the middle-aged P. gingivalis infected mice were increased. Conclusions These results suggest that P. gingivalis periodontal infection may cause cognitive impairment via the release of the pro-inflammatory cytokines TNF-α, IL-6, and IL-1β in the brain tissues of middle-aged mice.
Collapse
Affiliation(s)
- Ye Ding
- 1Department of Implantology, School and Hospital of Stomatology, Jilin University, Qinghua Road 1500, Chaoyang District, Changchun, 130021 China
| | - Jingyi Ren
- 1Department of Implantology, School and Hospital of Stomatology, Jilin University, Qinghua Road 1500, Chaoyang District, Changchun, 130021 China
| | - Hongqiang Yu
- 1Department of Implantology, School and Hospital of Stomatology, Jilin University, Qinghua Road 1500, Chaoyang District, Changchun, 130021 China
| | - Weixian Yu
- Key laboratory of Mechanism of Tooth Development and Jaw Bone Remodeling and Regeneration in Jilin Province, Qinghua Road 1500, Chaoyang District, Changchun, 130021 China
| | - Yanmin Zhou
- 1Department of Implantology, School and Hospital of Stomatology, Jilin University, Qinghua Road 1500, Chaoyang District, Changchun, 130021 China
| |
Collapse
|
45
|
Wang L, Wang H, Duan Z, Zhang J, Zhang W. Mechanism of gastrodin in cell apoptosis in rat hippocampus tissue induced by desflurane. Exp Ther Med 2018. [PMID: 29541166 PMCID: PMC5838295 DOI: 10.3892/etm.2018.5770] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
This study investigated the protective effect ofgastrodin on cell apoptosis in rats hippocampus tissues induced by desflurane to explore its mechanism. A total of 36 rats were randomly divided into three groups: Blank control group (C group, n=12), desflurane anesthesia group (DF group, n=12) and gastrodin treatment group (GT group, n=12). Rats in DF group were treated with anesthesia using desflurane. Rats in GT group were treated with gavage using gastrodin and the same treatment as DF group. After the experiment, novel object recognition test and water maze test were performed. The hippocampus tissues were taken from the rat after the behavioral experiment; then the number of apoptotic cells was detected using the terminal deoxynucleotidyltransferase-mediated dUTP nick end labelling (TUNEL) kit, and the mRNA and protein expression levels of p38 and interleukin-1 (IL-1) were detected via semi-quantitative polymerase chain reaction (PCR) and western blot analysis. After the desflurane anesthesia, novel object recognition showed that compared with that in DF group, the exploration capacity of novel objects in GT group was increased (P<0.01). The water maze test showed that the escape latencies in DF group, T1 in GT group was significantly shortened, but T2 was significantly prolonged (P<0.01). TUNEL assay showed that the number of apoptotic cells in hippocampus tissues in GT group was significantly fewer than that in group DF (P<0.01). Semi-quantitative PCR and western blot analysis showed that the expression levels of p38 and IL-1β in GT group were lower than those in DF group (P<0.01). The results show that gastrodin has a protective effect on the apoptosis of hippocampus cells of rats induced by desflurane. Its protection mechanism may be realized through decreasing the increased p38 and IL-1β expression levels induced by desflurane, thus blocking the p38 mitogen-activated protein kinase (p38 MAPK) pathway.
Collapse
Affiliation(s)
- Luping Wang
- Department of Anesthesiology, Hospital of Stomatology, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Hushan Wang
- Department of Anesthesiology, The First Bethune Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Zongsheng Duan
- Department of Anesthesiology, The First Bethune Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Jian Zhang
- Department of Anesthesiology, The First Bethune Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Wenwen Zhang
- Department of Anesthesiology, The First Bethune Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| |
Collapse
|
46
|
Safavynia SA, Goldstein PA. The Role of Neuroinflammation in Postoperative Cognitive Dysfunction: Moving From Hypothesis to Treatment. Front Psychiatry 2018; 9:752. [PMID: 30705643 PMCID: PMC6345198 DOI: 10.3389/fpsyt.2018.00752] [Citation(s) in RCA: 190] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 12/19/2018] [Indexed: 12/13/2022] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a common complication of the surgical experience and is common in the elderly and patients with preexisting neurocognitive disorders. Animal and human studies suggest that neuroinflammation from either surgery or anesthesia is a major contributor to the development of POCD. Moreover, a large and growing body of literature has focused on identifying potential risk factors for the development of POCD, as well as identifying candidate treatments based on the neuroinflammatory hypothesis. However, variability in animal models and clinical cohorts makes it difficult to interpret the results of such studies, and represents a barrier for the development of treatment options for POCD. Here, we present a broad topical review of the literature supporting the role of neuroinflammation in POCD. We provide an overview of the cellular and molecular mechanisms underlying the pathogenesis of POCD from pre-clinical and human studies. We offer a brief discussion of the ongoing debate on the root cause of POCD. We conclude with a list of current and hypothesized treatments for POCD, with a focus on recent and current human randomized clinical trials.
Collapse
Affiliation(s)
- Seyed A Safavynia
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY, United States
| | - Peter A Goldstein
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY, United States.,Department of Medicine, Weill Cornell Medical College, New York, NY, United States.,Neuroscience Graduate Program, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|