1
|
Wu Z, Yin Y, Liu R, Li X, Sun Y, Yau SY, Wu L, Liu Y, Adzic M, Zhang H, Chen G. A refined formula derived from Jiawei-Xiaoyao pill exerts rapid antidepressant-like effects in LPS-induced depression by reducing neuroinflammation and restoring neuroplasticity signaling. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118647. [PMID: 39094756 DOI: 10.1016/j.jep.2024.118647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/21/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jiawei-Xiaoyao Pill (JWX), a classic formula in traditional Chinese medicine, is derived from Xiaoyao Pill by adding significant amounts of Gardeniae Fructus (GF) and Moutan Cortex (MC). It is frequently used for the treatment of depression. JWX has been demonstrated to uniquely elicit rapid antidepressant-like effects within the prescribed dosage range. To date, GF has been shown to have rapid antidepressant-like effects, but a much higher dose is required than its proportion in JWX. It is assumed that the synergism of GF with a minimum number of other herbs in JWX serves as a refined formula that exerts these rapid antidepressant-like effects. Identification of a refined formula is important for prioritizing the herbs and ingredients to optimize the quality control of JWX. However, such a refined formula for JWX has not been identified yet. AIM OF THE STUDY Here we aimed to identify a refined formula derived from JWX for optimized rapid antidepressant-like effects. Since the neuroinflammation mechanism involving in depression treatment has not been previously investigated for JWX, we tested the mechanism for both JWX and the refined formula. MATERIALS AND METHODS Individual herbs (MC; ASR, Angelica Sinensis Radix; Bupleuri Radix; Paeonia Radix Alba) that show antidepressant-like responses were mixed with GF at the proportional dosage in JWX to identify the refined formula. Rapid antidepressant-like effects were assessed by using NSF (Novelty Suppressed Feeding Test) and other behavioral tests following a single administration. The identified formula was further tested in a lipopolysaccharide (LPS)-induced depressive model, and the molecular signaling mechanisms were investigated using Western blot analysis, immunofluorescence, and pharmacological inhibition of mTOR signaling. Scopolamine (Scop) was used as a positive control for induction of rapid antidepressant effects. RESULTS A combination of GF, MC and ASR (GMA) at their dosages proportional to JWX induced behavioral signs of rapid antidepressant-like responses in both normal and LPS-treated mice, with the antidepressant-like effects sustained for 5 d. Similar to JWX or Scop, GMA rapidly reduced the neuroinflammation signaling of Iba-1-NF-кB, enhanced neuroplasticity signaling of CaMKII-mTOR-BDNF, and attenuated the upregulated expressions of the NMDAR sub-units GluN1 and GluN2B in the hippocampus of LPS-treated mice. GMA, JWX and Scop rapidly restored the number of BDNF-positive cells reduced by LPS treatment in the CA3 region of the hippocampus. Furthermore, rapamycin, a selective inhibitor of mTOR, blunted the rapid antidepressant-like effects and hippocampal BDNF signaling upregulation by GMA. CONCLUSION GMA may serve as a refined formula from JWX, capable of inducing rapid antidepressant-like effects. In the LPS-induced depression model, the effects of GMA were mediated via rapidly alleviating neuroinflammation and enhancing neuroplasticity.
Collapse
Affiliation(s)
- Zhangjie Wu
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, PR China; Zhuhai Institute of Jinan University, Zhuhai, 519070, PR China; Guangdong-Hong Kong-Macao Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral Homeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, PR China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, PR China
| | - Ying Yin
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, PR China; Zhuhai Institute of Jinan University, Zhuhai, 519070, PR China; Guangdong-Hong Kong-Macao Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral Homeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, PR China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, PR China
| | - Ruiyi Liu
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, PR China; Zhuhai Institute of Jinan University, Zhuhai, 519070, PR China; Guangdong-Hong Kong-Macao Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral Homeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, PR China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, PR China
| | - Xianhui Li
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, PR China; Zhuhai Institute of Jinan University, Zhuhai, 519070, PR China; Guangdong-Hong Kong-Macao Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral Homeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, PR China
| | - Yan Sun
- Key Laboratory of Integrative Biomedicine for Brain Diseases, School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Suk-Yu Yau
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, PR China
| | - Lei Wu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, PR China
| | - Yan Liu
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, PR China; Zhuhai Institute of Jinan University, Zhuhai, 519070, PR China
| | - Miroslav Adzic
- "Vinča Institute" of Nuclear Sciences, Laboratory of Molecular Biology and Endocrinology 090, University of Belgrade, 11001 Belgrade, Serbia
| | - Hailou Zhang
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, PR China; Zhuhai Institute of Jinan University, Zhuhai, 519070, PR China; Guangdong-Hong Kong-Macao Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral Homeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, PR China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, PR China.
| | - Gang Chen
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, PR China; Zhuhai Institute of Jinan University, Zhuhai, 519070, PR China; Guangdong-Hong Kong-Macao Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral Homeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, PR China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, PR China.
| |
Collapse
|
2
|
Sherbini AHE, Hasheminia A, Gemae MR, Ansari F, Anood A, Saha T, Towe CW, El-Diasty M. Neuroinflammatory Pathways Associated with Chronic Post-Thoracotomy Pain: A Review of Current Literature. Mol Neurobiol 2024:10.1007/s12035-024-04565-y. [PMID: 39467985 DOI: 10.1007/s12035-024-04565-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
Chronic post-thoracotomy pain (CPTP) is a major clinical problem that affects up to 35-55% of patients undergoing thoracic incisions. Evidence suggests that multiple cellular signaling pathways and neuro-inflammatory mediators may play an essential role in the pathogenesis of CPTP. In this comprehensive review, we present the current evidence on the cellular signaling pathways and inflammatory changes associated with the initiation and maintenance of CPTP, focusing on the potential application of these findings in the clinical setting. An electronic search of Medline, EMBASE, Cochrane, Google Scholar, and ClinicalTrials.gov was performed, and 3652 abstracts were identified. After an initial abstract screening, 131 studies underwent a full-text review, and nine papers were eventually included in this review. Studies were included if they assessed the cellular signaling pathways or inflammatory processes associated with the induction and/or maintenance of CPTP. All the identified studies were pre-clinical studies conducted on animal models. Our search identified seven cellular pathways (NK-1 receptor (NK-1), Glutaminase 1, Toll-like receptor 4 (TLR4), Resolvins, Ror-2, Sonic hedgehog signaling (Shh), and Wnt5a/Wnts) and six cytokines (IL-1β, IL-6, IL-8, IL-10, IFN-γ, and TNF-α) that were investigated in the context of CPTP. Multiple cellular signaling pathways and inflammatory cytokines may play an important role in the neuroinflammatory changes associated with the induction and maintenance of chronic post-thoracotomy pain in animal models. However, the clinical impact and therapeutic utility of these neuroinflammatory changes in routine clinical practice have yet to be demonstrated.
Collapse
Affiliation(s)
| | - Amin Hasheminia
- Faculty of Health Sciences, Queen's University, Kingston, ON, Canada
| | - Mohamed R Gemae
- School of Medicine, Queen's University, Kingston, ON, Canada
| | - Farzan Ansari
- School of Medicine, Queen's University, Kingston, ON, Canada
| | - Alqaydi Anood
- Department of General Surgery, Queen's University, Kingston, ON, Canada
| | - Tarit Saha
- Department of Anesthesiology and Perioperative Medicine, Queen's University, Kingston, ON, Canada
| | - Christopher W Towe
- Division of Thoracic and Esophageal Surgery, Department of Surgery, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Mohammad El-Diasty
- Faculty of Health Sciences, Queen's University, Kingston, ON, Canada.
- Harrington Heart and Vascular Institute, Cardiac Surgery Department, University Hospitals Cleveland Medical Centre, Lakeside 3024, 11100 Euclid Ave, Cleveland, OH, 44106, USA.
| |
Collapse
|
3
|
Quintanilla B, Zarate CA, Pillai A. Ketamine's mechanism of action with an emphasis on neuroimmune regulation: can the complement system complement ketamine's antidepressant effects? Mol Psychiatry 2024; 29:2849-2858. [PMID: 38575806 DOI: 10.1038/s41380-024-02507-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 02/29/2024] [Indexed: 04/06/2024]
Abstract
Over 300 million people worldwide suffer from major depressive disorder (MDD). Unfortunately, only 30-40% of patients with MDD achieve complete remission after conventional monoamine antidepressant therapy. In recent years, ketamine has revolutionized the treatment of MDD, with its rapid antidepressant effects manifesting within a few hours as opposed to weeks with conventional antidepressants. Many research endeavors have sought to identify ketamine's mechanism of action in mood disorders; while many studies have focused on ketamine's role in glutamatergic modulation, several studies have implicated its role in regulating neuroinflammation. The complement system is an important component of the innate immune response vital for synaptic plasticity. The complement system has been implicated in the pathophysiology of depression, and studies have shown increases in complement component 3 (C3) expression in the prefrontal cortex of suicidal individuals with depression. Given the role of the complement system in depression, ketamine and the complement system's abilities to modulate glutamatergic transmission, and our current understanding of ketamine's anti-inflammatory properties, there is reason to suspect a common link between the complement system and ketamine's mechanism of action. This review will summarize ketamine's anti- inflammatory roles in the periphery and central nervous system, with an emphasis on complement system regulation.
Collapse
Affiliation(s)
- Brandi Quintanilla
- Pathophysiology of Neuropsychiatric Disorders Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Anilkumar Pillai
- Pathophysiology of Neuropsychiatric Disorders Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Research and Development, Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
4
|
De Jager JE, Boesjes R, Roelandt GHJ, Koliaki I, Sommer IEC, Schoevers RA, Nuninga JO. Shared effects of electroconvulsive shocks and ketamine on neuroplasticity: A systematic review of animal models of depression. Neurosci Biobehav Rev 2024; 164:105796. [PMID: 38981574 DOI: 10.1016/j.neubiorev.2024.105796] [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: 03/21/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/11/2024]
Abstract
Electroconvulsive shocks (ECS) and ketamine are antidepressant treatments with a relatively fast onset of therapeutic effects compared to conventional medication and psychotherapy. While the exact neurobiological mechanisms underlying the antidepressant response of ECS and ketamine are unknown, both interventions are associated with neuroplasticity. Restoration of neuroplasticity may be a shared mechanism underlying the antidepressant efficacy of these interventions. In this systematic review, literature of animal models of depression is summarized to examine the possible role of neuroplasticity in ECS and ketamine on a molecular, neuronal, synaptic and functional level, and specifically to what extent these mechanisms are shared between both interventions. The results highlight that hippocampal neurogenesis and brain-derived neurotrophic factor (BDNF) levels are consistently increased after ECS and ketamine. Moreover, both interventions positively affect glutamatergic neurotransmission, astrocyte and neuronal morphology, synaptic density, vasculature and functional plasticity. However, a small number of studies investigated these processes after ECS. Understanding the shared fundamental mechanisms of fast-acting antidepressants can contribute to the development of novel therapeutic approaches for patients with severe depression.
Collapse
Affiliation(s)
- Jesca E De Jager
- Department of Biomedical Sciences, Brain Center, University Medical Center, Groningen, the Netherlands.
| | - Rutger Boesjes
- University Centre of Psychiatry, University Medical Center Groningen, the Netherlands
| | - Gijs H J Roelandt
- University Centre of Psychiatry, University Medical Center Groningen, the Netherlands
| | - Ilektra Koliaki
- University Centre of Psychiatry, University Medical Center Groningen, the Netherlands
| | - Iris E C Sommer
- Department of Biomedical Sciences, Brain Center, University Medical Center, Groningen, the Netherlands
| | - Robert A Schoevers
- University Centre of Psychiatry, University Medical Center Groningen, the Netherlands
| | - Jasper O Nuninga
- Department of Biomedical Sciences, Brain Center, University Medical Center, Groningen, the Netherlands; University Medical Centre Utrecht, Department of Psychiatry, the Netherlands
| |
Collapse
|
5
|
Li S, Huang J, Luo D, Fu W, Liu J. Electro-acupuncture inhibits HDAC2 via modulating gut microbiota to ameliorate SNI-induced pain and depression-like behavior in rats. J Affect Disord 2024; 360:305-313. [PMID: 38395201 DOI: 10.1016/j.jad.2024.02.069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/02/2024] [Accepted: 02/18/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Depression and chronic pain frequent co-occur, exacerbating each other's symptoms and hindering treatment. Emerging studies have highlighted abnormal gut microbiota in both conditions. Previous studies have demonstrated the clinical effectiveness of electro-acupuncture (EA) in managing these conditions, yet the underlying mechanisms remain elusive. METHODS Spared nerve injury (SNI) was employed to induce chronic pain and depression-like behavior. Rats were randomly assigned to sham SNI (SS), SNI, and EA groups. SNI surgery was performed on all rats, except those in SS group, which underwent sham SNI surgery. Then EA group received 5 weeks of EA treatment. Pain and depression-like behavior were assessed through paw withdrawal threshold, sucrose-preference test, and forced swim test. Gut microbiota composition was analyzed via 16S rDNA sequencing. Brain-Derived Neurotrophic Factor (BDNF) and acetylation-related proteins in the medial prefrontal cortex (mPFC) were evaluated through enzyme-linked immunosorbent assay and western blot. RESULTS EA treatment significantly ameliorated pain and depression-like behavior. The 16S rDNA sequencing showed EA modulated gut microbiota composition, increased short-chain fatty acids (SCFAs)-producing bacteria, including Akkermansi, Ruminococcaceae and Lachnospiraceae family, particularly Akkermansia. Furthermore, EA increased BDNF, AcH3 and decreased HDAC2 in mPFC. Notably, SCFAs-producing bacteria exhibited a negative correlation with HDAC2 levels. LIMITATIONS This study exclusively investigated microbiota differences resulting from EA stimulation, without delving into the functional variations brought about by these microbial distinctions. CONCLUSIONS The therapeutic effects of EA on the comorbidity of chronic pain and depression may involve the modulation of the gut microbiota, resulting in histone acetylation changes and upregulation of BDNF.
Collapse
Affiliation(s)
- Sheng Li
- Department of Acupuncture and Moxibustion, The second affiliated hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Jianpeng Huang
- Department of Acupuncture and Moxibustion, The second affiliated hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ding Luo
- Department of Acupuncture and Moxibustion, The second affiliated hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wenbin Fu
- Department of Acupuncture and Moxibustion, The second affiliated hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Jianhua Liu
- Department of Acupuncture and Moxibustion, The second affiliated hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
6
|
Lin CL, Li YL, Chen YW, Kuo CH, Tu TY, Liu YF, Tsai JC, Shyong YJ. Amphiphilic NLC-Gel formulation loaded with Sebacoyl dinalbuphine ester and Nalbuphine for localized postoperative pain management. Int J Pharm 2024; 659:124295. [PMID: 38823469 DOI: 10.1016/j.ijpharm.2024.124295] [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: 02/26/2024] [Revised: 05/13/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
Opioids are powerful analgesics; however, their significant systemic adverse effects and the need for frequent administration restrict their use. Nalbuphine (NA) is a κ-agonist narcotic with limited adverse effects, but needs to be frequently administrated due to its short elimination half-life. Whereas sebacoyl dinalbuphine ester (SDE) is a NA prodrug, which can effectively prolong the analgesic effect, but lacks immediate pain relief. Therefore, in this study, a rapid and sustained local delivery formulation to introduce NA and SDE directly into surgical sites was developed. An amphiphilic nanostructured lipid carrier (NLC) poloxamer 407 (P407) gel (NLC-Gel) was developed to permit concurrent delivery of hydrophobic SDE from the NLC core and hydrophilic NA from P407, offering a dual rapid and prolonged analgesic effect. Benefiting from the thermal-sensitive characteristic of P407, the formulation can be injected in liquid phase and instantly transit into gel at wound site. NLC-Gel properties, including particle size, drug release, rheology, and stability, were assessed. In vivo evaluation using a rat spinal surgery model highlighted the effect of the formulation through pain behavior test and hematology analysis. NLC-Gels demonstrated an analgesic effect comparable with that of commercial intramuscular injected SDE formulation (IM SDE), with only 15 % of the drug dosage. The inclusion of supplemental NA in the exterior gel (PA12-Gel + NA) provided rapid drug onset owing to swift NA dispersion, addressing acute pain within hours along with prolonged analgesic effects. Our findings suggest that this amphiphilic formulation significantly enhanced postoperative pain management in terms of safety and efficacy.
Collapse
Affiliation(s)
- Cheng-Li Lin
- Department of Orthopedic Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Lian Li
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Wei Chen
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Cheng-Hsiang Kuo
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | - Ting-Yuan Tu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Yuan-Fu Liu
- Department of Orthopedic Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jui-Chen Tsai
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yan-Jye Shyong
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
7
|
Jiao J, Fan J, Zhang Y, Chen L. Efficacy and Safety of Ketamine to Treat Cancer Pain in Adult Patients: A Systematic Review. J Pain Symptom Manage 2024; 67:e185-e210. [PMID: 37972720 DOI: 10.1016/j.jpainsymman.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023]
Abstract
CONTEXT Ketamine is a well-characterized anesthetic agent, and subanesthetic ketamine possesses analgesic effects in both acute and chronic pain. OBJECTIVES A systematic review was performed to ascertain the efficacy and safety of ketamine in treating pain for cancer patients. METHODS Eight databases were searched from the inception to March 20th, 2023 to obtain randomized controlled trials (RCTs) on ketamine for treating pain in cancer patients. Two reviewers independently screened studies, extracted the data and assessed the risk of bias of included studies; then, meta-analysis was performed by using Revman 5.3 software and Stata 14.0 software. RESULTS Thirty-five studies were included, involving 2279 patients with cancer pain. The results of meta-analysis showed that ketamine could significantly reduce pain intensity. Subgroup analysis revealed that, when compared with control group, ketamine decreased markedly visual analogue scale (VAS) scores in two days after the end of treatment with ketamine, and ketamine administrated by patient controlled epidural analgesia (PCEA) was effective. Meanwhile, ketamine could significantly reduce the number of patient-controlled analgesia (PCA) compressions within 24 hours and morphine dosage. Ketamine could not decrease Ramsay sedation score. Additionally, the adverse events significantly decreased in the ketamine group, including nausea and vomiting, constipation, pruritus, lethargy, uroschesis, hallucination, and respiratory depression. In addition, compared with the control group, ketamine could reduce Hamilton depression scale (HAMD) score and relieve depressive symptoms. CONCLUSION Ketamine may be used as an effective therapy to relieve cancer pain. However, more rigorously designed RCTs with larger sample sizes are required to verify the above conclusions.
Collapse
Affiliation(s)
- Jiao Jiao
- Department of Anesthesiology (J.J., L.C.), West China Hospital, Sichuan University, Chengdu, China; The Research Units of West China-Chinese Academy of Medical Sciences (J.J., L.C.), West China Hospital, Sichuan University, Chengdu, China
| | - Jin Fan
- School of Acupuncture-Moxibustion and Tuina (J.F.), Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yonggang Zhang
- Department of Periodical Press and National Clinical Research Center for Geriatrics (Y.Z.), West China Hospital, Sichuan University, Chengdu, China; Chinese Evidence-based Medicine Center (Y.Z.), West China Hospital, Sichuan University, Chengdu, China; Nursing Key Laboratory of Sichuan Province (Y.Z.), Chengdu, China
| | - Lingmin Chen
- Department of Anesthesiology (J.J., L.C.), West China Hospital, Sichuan University, Chengdu, China; The Research Units of West China-Chinese Academy of Medical Sciences (J.J., L.C.), West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
8
|
Cui L, Li S, Wang S, Wu X, Liu Y, Yu W, Wang Y, Tang Y, Xia M, Li B. Major depressive disorder: hypothesis, mechanism, prevention and treatment. Signal Transduct Target Ther 2024; 9:30. [PMID: 38331979 PMCID: PMC10853571 DOI: 10.1038/s41392-024-01738-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/24/2023] [Accepted: 12/28/2023] [Indexed: 02/10/2024] Open
Abstract
Worldwide, the incidence of major depressive disorder (MDD) is increasing annually, resulting in greater economic and social burdens. Moreover, the pathological mechanisms of MDD and the mechanisms underlying the effects of pharmacological treatments for MDD are complex and unclear, and additional diagnostic and therapeutic strategies for MDD still are needed. The currently widely accepted theories of MDD pathogenesis include the neurotransmitter and receptor hypothesis, hypothalamic-pituitary-adrenal (HPA) axis hypothesis, cytokine hypothesis, neuroplasticity hypothesis and systemic influence hypothesis, but these hypothesis cannot completely explain the pathological mechanism of MDD. Even it is still hard to adopt only one hypothesis to completely reveal the pathogenesis of MDD, thus in recent years, great progress has been made in elucidating the roles of multiple organ interactions in the pathogenesis MDD and identifying novel therapeutic approaches and multitarget modulatory strategies, further revealing the disease features of MDD. Furthermore, some newly discovered potential pharmacological targets and newly studied antidepressants have attracted widespread attention, some reagents have even been approved for clinical treatment and some novel therapeutic methods such as phototherapy and acupuncture have been discovered to have effective improvement for the depressive symptoms. In this work, we comprehensively summarize the latest research on the pathogenesis and diagnosis of MDD, preventive approaches and therapeutic medicines, as well as the related clinical trials.
Collapse
Affiliation(s)
- Lulu Cui
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Shu Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Siman Wang
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Xiafang Wu
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Yingyu Liu
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Weiyang Yu
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Yijun Wang
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Yong Tang
- International Joint Research Centre on Purinergic Signalling/Key Laboratory of Acupuncture for Senile Disease (Chengdu University of TCM), Ministry of Education/School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine/Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China
| | - Maosheng Xia
- Department of Orthopaedics, The First Hospital, China Medical University, Shenyang, China.
| | - Baoman Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China.
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China.
- China Medical University Centre of Forensic Investigation, Shenyang, China.
| |
Collapse
|
9
|
Yu W, Wu Z, Li X, Ding M, Xu Y, Zhao P. Ketamine counteracts sevoflurane-induced depressive-like behavior and synaptic plasticity impairments through the adenosine A2A receptor/ERK pathway in rats. Mol Neurobiol 2023; 60:6160-6175. [PMID: 37428405 DOI: 10.1007/s12035-023-03474-w] [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: 12/28/2022] [Accepted: 07/02/2023] [Indexed: 07/11/2023]
Abstract
Ketamine is an ionic glutamic acid N-methyl-d-aspartate receptor (NMDAR) antagonist commonly used in clinical anesthesia, and its rapid and lasting antidepressant effect has stimulated great interest in psychology research. However, the molecular mechanisms underlying its antidepressant action are still undetermined. Sevoflurane exposure early in life might induce developmental neurotoxicity and mood disorders. In this study, we evaluated the effect of ketamine against sevoflurane-induced depressive-like behavior and the underlying molecular mechanisms. Here, we reported that A2AR protein expression was upregulated in rats with depression induced by sevoflurane inhalation, which was reversed by ketamine. Pharmacological experiments showed that A2AR agonists could reverse the antidepressant effect of ketamine, decrease extracellular signal-regulated kinase (ERK) phosphorylation, reduce synaptic plasticity, and induce depressive-like behavior. Our results suggest that ketamine mediates ERK1/2 phosphorylation by downregulating A2AR expression and that p-ERK1/2 increases the production of synaptic-associated proteins, enhancing synaptic plasticity in the hippocampus and thereby ameliorating the depressive-like behavior induced by sevoflurane inhalation in rats. This research provides a framework for reducing anesthesia-induced developmental neurotoxicity and developing new antidepressants.
Collapse
Affiliation(s)
- Weiwei Yu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Ziyi Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Xingyue Li
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Mengmeng Ding
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Ying Xu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Ping Zhao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China.
| |
Collapse
|
10
|
Wang YB, Song NN, Ding YQ, Zhang L. Neural plasticity and depression treatment. IBRO Neurosci Rep 2023; 14:160-184. [PMID: 37388497 PMCID: PMC10300479 DOI: 10.1016/j.ibneur.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/29/2022] [Accepted: 09/01/2022] [Indexed: 12/08/2022] Open
Abstract
Depression is one of the most common mental disorders, which can lead to a variety of emotional problems and even suicide at its worst. As this neuropsychiatric disorder causes the patients to suffer a lot and function poorly in everyday life, it is imposing a heavy burden on the affected families and the whole society. Several hypotheses have been proposed to elucidate the pathogenesis of depression, such as the genetic mutations, the monoamine hypothesis, the hypothalamic-pituitary-adrenal (HPA) axis hyperactivation, the inflammation and the neural plasticity changes. Among these models, neural plasticity can occur at multiple levels from brain regions, cells to synapses structurally and functionally during development and in adulthood. In this review, we summarize the recent progresses (especially in the last five years) on the neural plasticity changes in depression under different organizational levels and elaborate different treatments for depression by changing the neural plasticity. We hope that this review would shed light on the etiological studies for depression and on the development of novel treatments.
Collapse
Affiliation(s)
- Yu-Bing Wang
- Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center) and Department of Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai 200092, China
| | - Ning-Ning Song
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudfan University, Shanghai 200032, China
| | - Yu-Qiang Ding
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudfan University, Shanghai 200032, China
| | - Lei Zhang
- Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center) and Department of Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai 200092, China
| |
Collapse
|
11
|
Li Y, Shi W, Dai J, Jia Q, Guo G, Zhang Y, Zhang W. Upregulated TNF-α and lactate following ERK-SGK1 activation in the spinal dorsal horn underlies chronic postsurgical pain. CHINESE J PHYSIOL 2023; 66:144-152. [PMID: 37322625 DOI: 10.4103/cjop.cjop-d-22-00085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023] Open
Abstract
Skin/muscle incision and retraction (SMIR) during surgeries can lead to chronic postsurgical pain (CPSP). The underlying mechanisms are still unclear. In the present study, we showed that SMIR of the thigh induced phosphorylation of extracellular signal-regulated kinase (ERK), followed by serum- and glucocorticoid-inducible kinase-1 (SGK1) activation in the spinal dorsal horn. Intrathecal injection of PD98059, an ERK inhibitor, or GSK650394, a SGK1 inhibitor, significantly attenuated mechanical pain hypersensitivity in SMIR rats. The level of tumor necrosis factor α and lactate in spinal cord was significantly decreased by PD98059 or GSK650394 injection. Furthermore, PD98059 decreased the activation of SGK1 in the spinal dorsal horn. These results indicate that ERK-SGK1 activation followed by proinflammatory mediator release in the spinal dorsal horn underlies CPSP.
Collapse
Affiliation(s)
- Yuying Li
- School of Medical Technology and Nursing, Shenzhen Polytechnic, Shenzhen, Guangdong, China
| | - Wenjuan Shi
- Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Juanli Dai
- Department of Neurology, Xiehe Shenzhen Hospital, Huazhong University of Science and Technology, Shenzhen, Guangdong, China
| | - Qi Jia
- School of Medical Technology and Nursing, Shenzhen Polytechnic, Shenzhen, Guangdong, China
| | - Gang Guo
- School of Medical Technology and Nursing, Shenzhen Polytechnic, Shenzhen, Guangdong, China
| | | | - Weihong Zhang
- School of Medical Technology and Nursing, Shenzhen Polytechnic, Shenzhen, Guangdong, China
| |
Collapse
|
12
|
Serafini G, Costanza A, Aguglia A, Amerio A, Trabucco A, Escelsior A, Sher L, Amore M. The Role of Inflammation in the Pathophysiology of Depression and Suicidal Behavior: Implications for Treatment. Med Clin North Am 2023; 107:1-29. [PMID: 36402492 DOI: 10.1016/j.mcna.2022.09.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Depression and suicidal behavior are 2 complex psychiatric conditions of significant public health concerns due to their debilitating nature. The need to enhance contemporary treatments and preventative approaches for these illnesses not only calls for distillation of current views on their pathogenesis but also provides an impetus for further elucidation of their novel etiological determinants. In this regard, inflammation has recently been recognized as a potentially important contributor to the development of depression and suicidal behavior. This review highlights key evidence that supports the presence of dysregulated neurometabolic and immunologic signaling and abnormal interaction with microbial species as putative etiological hallmarks of inflammation in depression as well as their contribution to the development of suicidal behavior. Furthermore, therapeutic insights addressing candidate mechanisms of pathological inflammation in these disorders are proposed.
Collapse
Affiliation(s)
- Gianluca Serafini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health DINOGMI, Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, Genoa 16132, Italy.
| | - Alessandra Costanza
- Department of Psychiatry, Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland, Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Lugano, Switzerland; Department of Psychiatry, Faculty of Biomedical Sciences, University of Italian Switzerland (USI), Lugano, Switzerland
| | - Andrea Aguglia
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health DINOGMI, Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, Genoa 16132, Italy
| | - Andrea Amerio
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health DINOGMI, Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, Genoa 16132, Italy
| | - Alice Trabucco
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health DINOGMI, Section of Psychiatry, University of Genoa, Genoa, Italy
| | - Andrea Escelsior
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health DINOGMI, Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, Genoa 16132, Italy
| | - Leo Sher
- James J. Peters VA Medical Center, Bronx, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, New York, NY, USA
| | - Mario Amore
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health DINOGMI, Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, Genoa 16132, Italy
| |
Collapse
|
13
|
Cartágenes SDC, da Silveira CCSDM, Pinheiro BG, Fernandes LMP, Farias SV, Kobayashi NHC, de Souza PHFS, do Prado AF, Ferreira MKM, Lima RR, de Oliveira EHC, de Luna FCF, Burbano RMR, Fontes-Júnior EA, Maia CDSF. “K-Powder” Exposure during Adolescence Elicits Psychiatric Disturbances Associated with Oxidative Stress in Female Rats. Pharmaceuticals (Basel) 2022; 15:ph15111373. [PMID: 36355545 PMCID: PMC9698848 DOI: 10.3390/ph15111373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022] Open
Abstract
Ketamine, also called ‘K-powder’ by abusers, an analog of phencyclidine, primarily acts as an antagonist of N-methyl-D-aspartic acid (NMDA) receptors, therapeutically used as an anesthetic agent. Ketamine also stimulates the limbic system, inducing hallucinations and dissociative effects. At sub-anesthetic doses, ketamine also displays hallucinatory and dissociative properties, but not loss of consciousness. These behavioral consequences have elicited its recreational use worldwide, mainly at rave parties. Ketamine is generally a drug of choice among teenagers and young adults; however, the harmful consequences of its recreational use on adolescent central nervous systems are poorly explored. Thus, the aim of the present study was to characterize the behavioral and biochemical consequences induced by one binge-like cycle of ketamine during the early withdrawal period in adolescent female rats. Adolescent female Wistar rats (n = 20) received intraperitoneally administered ketamine (10 mg/kg/day) for 3 consecutive days. Twenty-four hours after the last administration of ketamine, animals were submitted to behavioral tests in an open field, elevated plus-maze, and forced swimming test. Then, animals were intranasally anesthetized with 2% isoflurane and euthanized to collect prefrontal cortex and hippocampus to assess lipid peroxidation, antioxidant capacity against peroxyl radicals, reactive oxygen species, reduced glutathione, and brain-derived neurotrophic factor (BDNF) levels. Our results found that 24 h after recreational ketamine use, emotional behavior disabilities, such as anxiety- and depression-like profiles, were detected. In addition, spontaneous ambulation was reduced. These negative behavioral phenotypes were associated with evidence of oxidative stress on the prefrontal cortex and hippocampus.
Collapse
Affiliation(s)
- Sabrina de Carvalho Cartágenes
- Laboratory of Pharmacology of Inflammation and Behavior, Health Sciences Institute, Pharmacy College, Federal University of Pará, Belém 66075-900, PA, Brazil
| | | | - Bruno Gonçalves Pinheiro
- Laboratory of Pharmacology of Inflammation and Behavior, Health Sciences Institute, Pharmacy College, Federal University of Pará, Belém 66075-900, PA, Brazil
| | - Luanna Melo Pereira Fernandes
- Laboratory of Pharmacology of Inflammation and Behavior, Health Sciences Institute, Pharmacy College, Federal University of Pará, Belém 66075-900, PA, Brazil
- Physiological and Morphological Sciences Department, Biological and Health Science Centre, State University of Pará, Belém 66087-662, PA, Brazil
| | - Sarah Viana Farias
- Laboratory of Pharmacology of Inflammation and Behavior, Health Sciences Institute, Pharmacy College, Federal University of Pará, Belém 66075-900, PA, Brazil
| | - Natália Harumi Correa Kobayashi
- Laboratory of Pharmacology of Inflammation and Behavior, Health Sciences Institute, Pharmacy College, Federal University of Pará, Belém 66075-900, PA, Brazil
| | - Pablo Henrique Franco Santos de Souza
- Laboratory of Pharmacology of Inflammation and Behavior, Health Sciences Institute, Pharmacy College, Federal University of Pará, Belém 66075-900, PA, Brazil
| | - Alejandro Ferraz do Prado
- Laboratory of Pharmacology and Toxicology of Cardiovascular System, Institute of Biological Science, Federal University of Pará, Belém 66075-900, PA, Brazil
| | - Maria Karolina Martins Ferreira
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-900, PA, Brazil
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-900, PA, Brazil
| | - Edivaldo Herculano Correa de Oliveira
- Laboratory of Cytogenomics and Environmental Mutagenesis, Environment Section (SAMAM), Evandro Chagas Institute (IEC), Ananindeua 67030-000, PA, Brazil
| | - Francisco Canindé Ferreira de Luna
- Laboratory of Cytogenomics and Environmental Mutagenesis, Environment Section (SAMAM), Evandro Chagas Institute (IEC), Ananindeua 67030-000, PA, Brazil
| | | | - Enéas Andrade Fontes-Júnior
- Laboratory of Pharmacology of Inflammation and Behavior, Health Sciences Institute, Pharmacy College, Federal University of Pará, Belém 66075-900, PA, Brazil
| | - Cristiane do Socorro Ferraz Maia
- Laboratory of Pharmacology of Inflammation and Behavior, Health Sciences Institute, Pharmacy College, Federal University of Pará, Belém 66075-900, PA, Brazil
- Correspondence:
| |
Collapse
|
14
|
Domínguez-Oliva A, Mota-Rojas D, Hernández-Avalos I, Mora-Medina P, Olmos-Hernández A, Verduzco-Mendoza A, Casas-Alvarado A, Whittaker AL. The neurobiology of pain and facial movements in rodents: Clinical applications and current research. Front Vet Sci 2022; 9:1016720. [PMID: 36246319 PMCID: PMC9556725 DOI: 10.3389/fvets.2022.1016720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022] Open
Abstract
One of the most controversial aspects of the use of animals in science is the production of pain. Pain is a central ethical concern. The activation of neural pathways involved in the pain response has physiological, endocrine, and behavioral consequences, that can affect both the health and welfare of the animals, as well as the validity of research. The strategy to prevent these consequences requires understanding of the nociception process, pain itself, and how assessment can be performed using validated, non-invasive methods. The study of facial expressions related to pain has undergone considerable study with the finding that certain movements of the facial muscles (called facial action units) are associated with the presence and intensity of pain. This review, focused on rodents, discusses the neurobiology of facial expressions, clinical applications, and current research designed to better understand pain and the nociceptive pathway as a strategy for implementing refinement in biomedical research.
Collapse
Affiliation(s)
- Adriana Domínguez-Oliva
- Master in Science Program “Maestría en Ciencias Agropecuarias”, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Daniel Mota-Rojas
- Neurophysiology, Behavior and Animal Welfare Assesment, DPAA, Universidad Autónoma Metropolitana, Mexico City, Mexico
- *Correspondence: Daniel Mota-Rojas
| | - Ismael Hernández-Avalos
- Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli, Mexico
| | - Patricia Mora-Medina
- Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli, Mexico
| | - Adriana Olmos-Hernández
- Division of Biotechnology-Bioterio and Experimental Surgery, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| | - Antonio Verduzco-Mendoza
- Division of Biotechnology-Bioterio and Experimental Surgery, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| | - Alejandro Casas-Alvarado
- Neurophysiology, Behavior and Animal Welfare Assesment, DPAA, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Alexandra L. Whittaker
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| |
Collapse
|
15
|
Zhao L, Zhang H, Cheng H. Effect of a single sub-dose of ketamine on postoperative fatigue syndrome in colorectal cancer patients undergoing radical laparoscopic surgery: A double-blind, pilot study. J Affect Disord 2022; 312:146-151. [PMID: 35750094 DOI: 10.1016/j.jad.2022.06.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 06/12/2022] [Accepted: 06/17/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND This study aimed at examining the effect of a single sub-anesthetic dose of ketamine on postoperative fatigue syndrome (POFS) in patients undergoing radical laparoscopic surgery for colorectal cancer (CRC). METHODS This prospective, double-blind, pilot study enrolled patients scheduled for radical laparoscopic surgery for CRC under general anesthesia. Eligible patients were randomized into the placebo and ketamine groups. The primary outcome was christensen score change at day 3. The secondary outcomes were the difference of Identity Consequence Fatigue Scale (ICFS) score between the placebo group and ketamine group at day 3 and level of serum tumor necrosis factor (TNF)-α, interleukin (IL)-6, S100β protein, and neuron-specific enolase (NSE). RESULTS 32 participants were assigned to the ketamine group and 31 to the placebo group. Compared with placebo group, the Christensen score was lower in ketamine group at day 3 (absolute difference, -1.13; 95 % confidence interval [CI], -2.02 to -0.24; P = 0.012). Ketamine group was superior to the placebo group with regard to the ICFS scores at day 3 (absolute difference, -6.4; 95 % CI, -11.4 to -1.4; P = 0.013). The plasma TNF-α, IL-6, S100β, and NSE levels were increased after operation compared with baseline in both groups and were significantly higher in placebo group than in ketamine group within 24 h after surgery (all P < 0.05). There was no significant difference of each safety evaluation indicator between the two groups (all P > 0.05). CONCLUSION A single sub-anesthetic dose of ketamine may improve POFS in patients undergoing radical laparoscopic surgery for CRC, without postoperative adverse reactions.
Collapse
Affiliation(s)
- Liqin Zhao
- Department of Anesthesiology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Hailong Zhang
- Department of Anesthesiology, Beijing Luhe Hospital, Capital Medical University, Beijing, China.
| | - Hao Cheng
- Department of Anesthesiology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
16
|
Liu J, Yu E. P2RY12 Increased Neuroinflammation to Accelerate Depression-like Behaviors by the NLPR3 Inflammasome. Curr Neurovasc Res 2022; 19:267-274. [PMID: 36043776 DOI: 10.2174/1567202619666220829110111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/10/2022] [Accepted: 07/20/2022] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Depression is a class of important mental illness, which has become a severe health problem perplexing the world due to its high morbidity rate, high disability rate, and great disease burden. This study aimed to evaluate the role and possible mechanisms of P2RY12 in the depression-like behaviors model. METHODS Serum samples of patients with depression-like behaviors were used to analyze the expression of P2RY12. Models of mice were given LPS via intraperitoneal injection for 7 days. Behavioral tests were executed in this experiment. RESULTS The expression of P2RY12 in models of depression-like behaviors or mice with depression- like behaviors were induced. The inhibition of P2RY12 presents depression-like behaviors and reduces inflammation in the model of depression-like behaviors. P2RY12 induced NLRP3 expression and suppressed NLRP3 ubiquitination in a model of depression-like behavior. The inhibition of NLRP3 reduced the effects of P2RY12 in mice model of depression-like behaviors. The regulation of NLRP3 controlled the effects of the P2RY12 in vitro model of depression-like behaviors. CONCLUSION We conclude that P2RY12 increased neuroinflammation to accelerate depression-like behaviors by NLPR3 inflammasome, providing novel information for the treatment of depressionlike behaviors.
Collapse
Affiliation(s)
- Jie Liu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hanzhou 310053, China
| | - Enyan Yu
- Department of Clinical Psychology, Zhejiang Cancer Hospital, Hanzhou 310012, China
| |
Collapse
|
17
|
Dysfunctional Heteroreceptor Complexes as Novel Targets for the Treatment of Major Depressive and Anxiety Disorders. Cells 2022; 11:cells11111826. [PMID: 35681521 PMCID: PMC9180493 DOI: 10.3390/cells11111826] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/10/2022] [Accepted: 05/20/2022] [Indexed: 02/01/2023] Open
Abstract
Among mental diseases, major depressive disorder (MDD) and anxiety deserve a special place due to their high prevalence and their negative impact both on society and patients suffering from these disorders. Consequently, the development of novel strategies designed to treat them quickly and efficiently, without or at least having limited side effects, is considered a highly important goal. Growing evidence indicates that emerging properties are developed on recognition, trafficking, and signaling of G-protein coupled receptors (GPCRs) upon their heteromerization with other types of GPCRs, receptor tyrosine kinases, and ionotropic receptors such as N-methyl-D-aspartate (NMDA) receptors. Therefore, to develop new treatments for MDD and anxiety, it will be important to identify the most vulnerable heteroreceptor complexes involved in MDD and anxiety. This review focuses on how GPCRs, especially serotonin, dopamine, galanin, and opioid heteroreceptor complexes, modulate synaptic and volume transmission in the limbic networks of the brain. We attempt to provide information showing how these emerging concepts can contribute to finding new ways to treat both MDD and anxiety disorders.
Collapse
|
18
|
Ketamine administration ameliorates anesthesia and surgery‑induced cognitive dysfunction via activation of TRPV4 channel opening. Exp Ther Med 2022; 24:478. [PMID: 35761804 PMCID: PMC9214599 DOI: 10.3892/etm.2022.11405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/16/2022] [Indexed: 11/05/2022] Open
|
19
|
Liu Y, Kuai S, Ding M, Wang Z, Zhao L, Zhao P. Dexmedetomidine and Ketamine Attenuated Neuropathic Pain Related Behaviors via STING Pathway to Induce ER-Phagy. Front Synaptic Neurosci 2022; 14:891803. [PMID: 35645765 PMCID: PMC9136071 DOI: 10.3389/fnsyn.2022.891803] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
Our previous work indicated that ER-phagy level had altered in spinal nerve ligation (SNL) rats. In this study, we investigated whether dexmedetomidine or ketamine exhibits anti-anxiety or anti-nociceptive effects via modulation of the spinal STING/TBK pathway to alter ER-phagy in SNL rats. We evaluated the analgesic and anti-anxiety effects of ketamine and dexmedetomidine in SNL rats. 2’3’-cGAMP (a STING pathway agonist) was administrated to investigate whether enhanced spinal STING pathway activation could inhibit dexmedetomidine or ketamine treatment effects in SNL rats. Analgesic effects were assessed with the mechanical withdrawal threshold (MWT) and anti-anxiety effects were measured via an open field test (OFT). Protein expression levels were evaluated by immunoblotting. Distribution and cellular localization of Grp78 (ER stress marker) were evaluated by confocal immunofluorescence. SNL induced mechanical hypersensitivity and anxiety in rats; dexmedetomidine and ketamine both provided analgesia and anti-anxiety effects in SNL rats. Furthermore, the STING pathway was involved in the modulation of ER stress and ER-phagy in SNL rats and dexmedetomidine and ketamine alleviated ER stress by inhibiting STING pathway to enhance ER-phagy. Thus, both ketamine and dexmedetomidine provided anti-anxiety and anti-nociceptive effects by alleviating ER stress through the inhibition of the STING/TBK pathway to modulate spinal ER-phagy in SNL rats.
Collapse
Affiliation(s)
- Yongda Liu
- Department of Anesthesiology and Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shihui Kuai
- Department of Anesthesiology and Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mengmeng Ding
- Department of Anesthesiology and Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhibin Wang
- Department of Anesthesiology and Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Limei Zhao
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Limei Zhao Ping Zhao
| | - Ping Zhao
- Department of Anesthesiology and Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Limei Zhao Ping Zhao
| |
Collapse
|
20
|
Riveros ME, Ávila A, Schruers K, Ezquer F. Antioxidant Biomolecules and Their Potential for the Treatment of Difficult-to-Treat Depression and Conventional Treatment-Resistant Depression. Antioxidants (Basel) 2022; 11:540. [PMID: 35326190 PMCID: PMC8944633 DOI: 10.3390/antiox11030540] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 11/23/2022] Open
Abstract
Major depression is a devastating disease affecting an increasing number of people from a young age worldwide, a situation that is expected to be worsened by the COVID-19 pandemic. New approaches for the treatment of this disease are urgently needed since available treatments are not effective for all patients, take a long time to produce an effect, and are not well-tolerated in many cases; moreover, they are not safe for all patients. There is solid evidence showing that the antioxidant capacity is lower and the oxidative damage is higher in the brains of depressed patients as compared with healthy controls. Mitochondrial disfunction is associated with depression and other neuropsychiatric disorders, and this dysfunction can be an important source of oxidative damage. Additionally, neuroinflammation that is commonly present in the brain of depressive patients highly contributes to the generation of reactive oxygen species (ROS). There is evidence showing that pro-inflammatory diets can increase depression risk; on the contrary, an anti-inflammatory diet such as the Mediterranean diet can decrease it. Therefore, it is interesting to evaluate the possible role of plant-derived antioxidants in depression treatment and prevention as well as other biomolecules with high antioxidant and anti-inflammatory potential such as the molecules paracrinely secreted by mesenchymal stem cells. In this review, we evaluated the preclinical and clinical evidence showing the potential effects of different antioxidant and anti-inflammatory biomolecules as antidepressants, with a focus on difficult-to-treat depression and conventional treatment-resistant depression.
Collapse
Affiliation(s)
- María Eugenia Riveros
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Santiago 7710162, Chile
| | - Alba Ávila
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Santiago 7710162, Chile;
| | - Koen Schruers
- Department of Psychiatry and Neuropsychology, Maastricht University Medical Center, 6229 Maastricht, The Netherlands;
| | - Fernando Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Santiago 7710162, Chile;
| |
Collapse
|
21
|
Ketamine inhibits TNF-α-induced cecal damage by enhancing RIP1 ubiquitination to attenuate lethal SIRS. Cell Death Dis 2022; 8:72. [PMID: 35184141 PMCID: PMC8857635 DOI: 10.1038/s41420-022-00869-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/22/2022] [Accepted: 02/03/2022] [Indexed: 01/02/2023]
Abstract
Systemic inflammatory response syndrome (SIRS) is a sepsis-associated inflammatory state and a self-defense mechanism against specific and nonspecific stimuli. Ketamine influences many key processes that are altered during sepsis. However, the underlying mechanisms remain incompletely understood. In this study, TNF-α-treated mice, as well as HT-29 and L929 cell models, were applied to characterize TNF-α-induced systemic and local cecal tissue inflammatory responses. Behavioral, biochemical, histological, and molecular biological approaches were applied to illustrate the related processes. Mice with TNF-α-induced SIRS showed systemic and local cecal tissue inflammatory responses, as indicated by increased levels of high mobility group box 1 protein (HMGB1), chemokines (C-X-C motif) ligand 10 (CXCL10), interleukin-6 (IL-6), and IL-10, as well as high mortality. Ketamine pretreatment alleviated death rates, symptoms, and the production of inflammatory cytokines induced by TNF-α in mice. Moreover, ketamine also protected the mice from TNF-α-induced cecal damage by suppressing the phosphorylation of receptor-interacting serine/threonine-protein kinase 3 (RIP3) and mixed lineage kinase domain-like protein (MLKL). In addition, our results showed that ketamine efficiently inhibited TNF-α-induced necroptosis in HT-29 and L929 cells. Furthermore, we explored the mechanism using different L929 cell lines. The results displayed that ketamine inhibited TNF-α-induced necroptosis by enhancing RIP1 ubiquitination and reducing the RIP1-RIP3 and RIP3-MLKL interactions, as well as the formation of necrosomes. Thus, our study may provide a new theoretical and experimental basis for treating diseases characterized by SIRS-associated inflammatory factor storms. Moreover, our exploration may provide potential molecular mechanisms and targets for therapeutic intervention and clinical application of ketamine.
Collapse
|
22
|
Porter GA, O’Connor JC. Brain-derived neurotrophic factor and inflammation in depression: Pathogenic partners in crime? World J Psychiatry 2022; 12:77-97. [PMID: 35111580 PMCID: PMC8783167 DOI: 10.5498/wjp.v12.i1.77] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/21/2021] [Accepted: 12/02/2021] [Indexed: 02/06/2023] Open
Abstract
Major depressive disorder is a debilitating disorder affecting millions of people each year. Brain-derived neurotrophic factor (BDNF) and inflammation are two prominent biologic risk factors in the pathogenesis of depression that have received considerable attention. Many clinical and animal studies have highlighted associations between low levels of BDNF or high levels of inflammatory markers and the development of behavioral symptoms of depression. However, less is known about potential interaction between BDNF and inflammation, particularly within the central nervous system. Emerging evidence suggests that there is bidirectional regulation between these factors with important implications for the development of depressive symptoms and anti-depressant response. Elevated levels of inflammatory mediators have been shown to reduce expression of BDNF, and BDNF may play an important negative regulatory role on inflammation within the brain. Understanding this interaction more fully within the context of neuropsychiatric disease is important for both developing a fuller understanding of biological pathogenesis of depression and for identifying novel therapeutic opportunities. Here we review these two prominent risk factors for depression with a particular focus on pathogenic implications of their interaction.
Collapse
Affiliation(s)
- Grace A Porter
- Department of Pharmacology, UT Health San Antonio, San Antonio, TX 78229, United States
| | - Jason C O’Connor
- Department of Pharmacology, University of Texas Health San Antonio, San Antonio, TX 78229, United States
- Audie L. Murphy VA Hospital, South Texas Veterans Health System, San Antonio, TX 78229, United States
| |
Collapse
|
23
|
Beating Pain with Psychedelics: Matter over Mind? Neurosci Biobehav Rev 2021; 134:104482. [PMID: 34922987 DOI: 10.1016/j.neubiorev.2021.12.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/19/2021] [Accepted: 12/04/2021] [Indexed: 02/08/2023]
Abstract
Basic pain research has shed light on key cellular and molecular mechanisms underlying nociceptive and phenomenological aspects of pain. Despite these advances, [[we still yearn for] the discovery of novel therapeutic strategies to address the unmet needs of about 70% of chronic neuropathic pain patients whose pain fails to respond to opioids as well as to other conventional analgesic agents. Importantly, a substantial body of clinical observations over the past decade cumulatively suggests that the psychedelic class of drugs may possess heuristic value for understanding and treating chronic pain conditions. The present review presents a theoretical framework for hitherto insufficiently understood neuroscience-based mechanisms of psychedelics' potential analgesic effects. To that end, searches of PubMed-indexed journals were performed using the following Medical Subject Headings' terms: pain, analgesia, inflammatory, brain connectivity, ketamine, psilocybin, functional imaging, and dendrites. Recursive sets of scientific and clinical evidence extracted from this literature review were summarized within the following key areas: (1) studies employing psychedelics for alleviation of physical and emotional pain; (2) potential neuro-restorative effects of psychedelics to remediate the impaired connectivity underlying the dissociation between pain-related conscious states/cognitions and the subcortical activity/function leading to the eventual chronicity through immediate and long-term effects on dentritic plasticity; (3) anti-neuroinflammatory and pro-immunomodulatory actions of psychedelics as the may pertain to the role of these factors in the pathogenesis of neuropathic pain; (4) safety, legal, and ethical consideration inherent in psychedelics' pharmacotherapy. In addition to direct beneficial effects in terms of reduction of pain and suffering, psychedelics' inclusion in the analgesic armamentarium will contribute to deeper and more sophisticated insights not only into pain syndromes but also into frequently comorbid psychiatric condition associated with emotional pain, e.g., depressive and anxiety disorders. Further inquiry is clearly warranted into the above areas that have potential to evolve into further elucidate the mechanisms of chronic pain and affective disorders, and lead to the development of innovative, safe, and more efficacious neurobiologically-based therapeutic approaches.
Collapse
|
24
|
Lu Y, Hu B, Dai H, Wang B, Yao J, Yao X. Predictors of Chronic Postsurgical Pain in Elderly Patients Undergoing Hip Arthroplasty: A Multi-Center Retrospective Cohort Study. Int J Gen Med 2021; 14:7885-7894. [PMID: 34785937 PMCID: PMC8591111 DOI: 10.2147/ijgm.s337170] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/26/2021] [Indexed: 12/16/2022] Open
Abstract
Background Identifying patients at high risk of developing chronic postsurgical pain (CPSP) is of extreme importance in order to help launch appropriate therapeutic strategies and intensive initiation of pain management. Aim In this study, we aimed to conduct a multi-center retrospective cohort study to establish a prognostic model and a nomogram to predict the risks of CPSP in elderly patients who underwent hip arthroplasty at 6 months postoperatively. Methods We collected data from 736 patients aged >65 years who had undergone hip arthroplasty from October 1, 2016 to September, 30, 2018 at multiple tertiary referral centers in Guangzhou, China. All data were randomly stratified into a training set and a testing set at a ratio of 8:2. Data were analyzed via multiple logistic regression analysis with receiver operating characteristic (ROC) curves and areas under the curve. This model was further validated by estimating calibration and discrimination. A nomogram was ultimately developed. Results A total of 736 eligible patients were enrolled, 27.20% of whom developed CPSP within 6 months postoperatively. Preoperative pain in the surgical area (OR=2.456, 95% CI:1.814–3.327, P<0.001), preoperative depression state (OR=1.256, 95% CI:1.146–1.378, P<0.001), surgical type (OR=7.138, 95% CI:3.548–14.364, P<0.001), acute postoperative numerical rating scale score (OR=5.537, 95% CI:3.607–8.499, P<0.001) and analgesic type (patient-controlled epidural analgesia: OR=0.129, 95% CI:0.055–0.299, P<0.001; patient-controlled intravenous analgesia: OR=0.033, 95% CI:0.011–0.097, P<0.001) were identified as independent significant factors associated with CPSP. A prognostic model was established and further validated. An ROC curve confirmed the predictive ability of this model with a high sensitivity value of 92.12% (95% CI:86.90–95.74) and specificity value of 91.72% (95% CI:88.77–94.11). A nomogram was developed to simplify the use of the predictive model in clinical practice. Conclusion This prognostic model could be of great value in clinical practice, serving as the basis for early personalized analgesic management of elderly patients undergoing hip arthroplasty. ![]()
Point your SmartPhone at the code above. If you have a QR code reader the video abstract will appear. Or use: https://youtu.be/Im3bJ1sYwM4
Collapse
Affiliation(s)
- Yi Lu
- Department of Anesthesiology, The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Bang Hu
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, People's Republic of China
| | - Haitao Dai
- Department of Interventional Radiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, People's Republic of China
| | - Bao Wang
- Department of Anesthesiology, The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Jiayin Yao
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, People's Republic of China
| | - Xinhua Yao
- Department of Anesthesiology, The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, People's Republic of China
| |
Collapse
|
25
|
Prowse N, Hayley S. Microglia and BDNF at the crossroads of stressor related disorders: Towards a unique trophic phenotype. Neurosci Biobehav Rev 2021; 131:135-163. [PMID: 34537262 DOI: 10.1016/j.neubiorev.2021.09.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/16/2022]
Abstract
Stressors ranging from psychogenic/social to neurogenic/injury to systemic/microbial can impact microglial inflammatory processes, but less is known regarding their effects on trophic properties of microglia. Recent studies do suggest that microglia can modulate neuronal plasticity, possibly through brain derived neurotrophic factor (BDNF). This is particularly important given the link between BDNF and neuropsychiatric and neurodegenerative pathology. We posit that certain activated states of microglia play a role in maintaining the delicate balance of BDNF release onto neuronal synapses. This focused review will address how different "activators" influence the expression and release of microglial BDNF and address the question of tropomyosin receptor kinase B (TrkB) expression on microglia. We will then assess sex-based differences in microglial function and BDNF expression, and how microglia are involved in the stress response and related disorders such as depression. Drawing on research from a variety of other disorders, we will highlight challenges and opportunities for modulators that can shift microglia to a "trophic" phenotype with a view to potential therapeutics relevant for stressor-related disorders.
Collapse
Affiliation(s)
- Natalie Prowse
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada.
| | - Shawn Hayley
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada.
| |
Collapse
|
26
|
Kopra E, Mondelli V, Pariante C, Nikkheslat N. Ketamine's effect on inflammation and kynurenine pathway in depression: A systematic review. J Psychopharmacol 2021; 35:934-945. [PMID: 34180293 PMCID: PMC8358579 DOI: 10.1177/02698811211026426] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Ketamine is a novel rapid-acting antidepressant with high efficacy in treatment-resistant patients. Its exact therapeutic mechanisms of action are unclear; however, in recent years its anti-inflammatory properties and subsequent downstream effects on tryptophan (TRP) metabolism have sparked research interest. AIM This systematic review examined the effect of ketamine on inflammatory markers and TRP-kynurenine (KYN) pathway metabolites in patients with unipolar and bipolar depression and in animal models of depression. METHODS MEDLINE, Embase, and PsycINFO databases were searched on October 2020 (1806 to 2020). RESULTS Out of 807 initial results, nine human studies and 22 animal studies on rodents met the inclusion criteria. Rodent studies provided strong support for ketamine-induced decreases in pro-inflammatory cytokines, namely in interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α and indicated anti-inflammatory effects on TRP metabolism, including decreases in the enzyme indoleamine 2,3-dioxygenase (IDO). Clinical evidence was less robust with high heterogeneity between sample characteristics, but most experiments demonstrated decreases in peripheral inflammation including in IL-1β, IL-6, and TNF-α. Preliminary support was also found for reduced activation of the neurotoxic arm of the KYN pathway. CONCLUSION Ketamine appears to induce anti-inflammatory effects in at least a proportion of depressed patients. Suggestions for future research include investigation of markers in the central nervous system and examination of clinical relevance of inflammatory changes.
Collapse
Affiliation(s)
- Emma Kopra
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Valeria Mondelli
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College 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, UK
| | - Carmine Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College 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, UK
| | - Naghmeh Nikkheslat
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| |
Collapse
|
27
|
Cui M, Dai W, Kong J, Chen H. Th17 Cells in Depression: Are They Crucial for the Antidepressant Effect of Ketamine? Front Pharmacol 2021; 12:649144. [PMID: 33935753 PMCID: PMC8082246 DOI: 10.3389/fphar.2021.649144] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/16/2021] [Indexed: 01/08/2023] Open
Abstract
Background: Major depressive disorder is associated with inflammation and immune processes. Depressive symptoms correlate with inflammatory markers and alterations in the immune system including cytokine levels and immune cell function. Th17 cells are a T cell subset which exerts proinflammatory effects. Th17 cell accumulation and Th17/Treg imbalances have been reported to be critical in the pathophysiology of major depressive disorder and depressive-like behaviors in animal models. Th17 cells are thought to interfere with glutamate signaling, dopamine production, and other immune processes. Ketamine is a newly characterized antidepressant medication which has proved to be effective in rapidly reducing depressive symptoms. However, the mechanisms behind these antidepressant effects have not been fully elucidated. Method: Literature about Th17 cells and their role in depression and the antidepressant effect of ketamine are reviewed, with the possible interaction networks discussed. Result: The immune-modulating role of Th17 cells may participate in the antidepressant effect of ketamine. Conclusion: As Th17 cells play multiple roles in depression, it is important to explore the mechanisms of action of ketamine on Th17 cells and Th17/Treg cell balance. This provides new perspectives for strengthening the antidepressant effect of ketamine while reducing its side effects and adverse reactions.
Collapse
Affiliation(s)
- Meiying Cui
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Wanlin Dai
- Innovation Institute of China Medical University, Shenyang, China
| | - Jing Kong
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hongzhi Chen
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| |
Collapse
|
28
|
Jansen van Vuren E, Steyn SF, Brink CB, Möller M, Viljoen FP, Harvey BH. The neuropsychiatric manifestations of COVID-19: Interactions with psychiatric illness and pharmacological treatment. Biomed Pharmacother 2021; 135:111200. [PMID: 33421734 PMCID: PMC7834135 DOI: 10.1016/j.biopha.2020.111200] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/15/2020] [Accepted: 12/26/2020] [Indexed: 12/12/2022] Open
Abstract
The recent outbreak of the corona virus disease (COVID-19) has had major global impact. The relationship between severe acute respiratory syndrome coronavirus (SARS-CoV-2) infection and psychiatric diseases is of great concern, with an evident link between corona virus infections and various central and peripheral nervous system manifestations. Unmitigated neuro-inflammation has been noted to underlie not only the severe respiratory complications of the disease but is also present in a range of neuro-psychiatric illnesses. Several neurological and psychiatric disorders are characterized by immune-inflammatory states, while treatments for these disorders have distinct anti-inflammatory properties and effects. With inflammation being a common contributing factor in SARS-CoV-2, as well as psychiatric disorders, treatment of either condition may affect disease progression of the other or alter response to pharmacological treatment. In this review, we elucidate how viral infections could affect pre-existing psychiatric conditions and how pharmacological treatments of these conditions may affect overall progress and outcome in the treatment of SARS-CoV-2. We address whether any treatment-induced benefits and potential adverse effects may ultimately affect the overall treatment approach, considering the underlying dysregulated neuro-inflammatory processes and potential drug interactions. Finally, we suggest adjunctive treatment options for SARS-CoV-2-associated neuro-psychiatric symptoms.
Collapse
Affiliation(s)
- Esmé Jansen van Vuren
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa.
| | - Stephan F Steyn
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Christiaan B Brink
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Marisa Möller
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Francois P Viljoen
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Brian H Harvey
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa; South African MRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|