1
|
Sakita M, Isobe W, Nonaka K, Murakami S, Miyachi R, Sakane K, Sugimoto S, Yamaguchi A, Yamamoto K. Age‑related changes in endoplasmic reticulum stress response‑associated protein expression in rat tibial nerves. Biomed Rep 2025; 22:50. [PMID: 39882333 PMCID: PMC11775640 DOI: 10.3892/br.2025.1928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025] Open
Abstract
In age-related peripheral neurodegeneration, changes in the promotion or inhibition of endoplasmic reticulum (ER) stress response related to the ubiquitin-proteasome degradation system (UPS), autophagy and apoptosis signaling factors during aging remain unclear. In the present study, the expression of ER stress response signaling-related protein factors was examined in tibial nerves during aging in rats. Tibial nerves were extracted from continuously housed rats at 20, 50, 70, 90 and 105 weeks of age. Expression of factors associated with ER stress-related degradation, including X-box binding protein 1 (XBP1s), eukaryotic translation initiation factor 2 subunit 1 (eIF2α), Beclin-1 (Becn1), and Caspase-3 (Casp3); ER stress-related repair, including binding immunoglobulin protein [also known as 78 kDa glucose-regulated protein (BiP/GRP78)], protein disulfide isomerase (PDI), brain-derived neurotrophic factor (BDNF) and the inflammatory cytokine IL6, was assessed by western blotting of tibial nerves from rats in each age group. Expression of XBP1s and Becn1, which promote UPS and autophagy, decreased significantly after 50 weeks of age. However, expression of eIF2α and Casp3, which inhibit new protein biosynthesis and promote apoptosis, increased significantly after 50 weeks. Expression of BiP/GRP78 and PDI, which are refolding factors for denatured proteins, showed a significant decrease after 50 (or 70) weeks of age. The expression of BDNF, a ligand protein for the repair cascade, showed a significant increase after 70 weeks of age, while that of IL6 increased significantly after 50 weeks of age. These results indicate that ER stress-related degradation (UPS and autophagy) and refolding repair functions are reduced in rat tibial nerves after 50 weeks, followed by enhanced apoptosis and inflammation. These findings shed light on the progression of age-related peripheral neurodegeneration in rats.
Collapse
Affiliation(s)
- Masahiro Sakita
- Department of Physical Therapy, Faculty of Health Sciences, Kyoto Tachibana University, Kyoto 607-8175, Japan
| | - Wataru Isobe
- Department of Physical Therapy, Faculty of Health Sciences, Kyoto Tachibana University, Kyoto 607-8175, Japan
- Department of Rehabilitation, Mitsubishi Kyoto Hospital, Kyoto 615-8087, Japan
| | - Koji Nonaka
- Department of Rehabilitation, Faculty of Health Care Sciences, Naragakuen University, Nara 631-0003, Japan
| | - Shinichiro Murakami
- Department of Physical Therapy, Faculty of Health Care Sciences, Himeji-Dokkyo University, Himeji, Hyogo 670-0896, Japan
| | - Ryo Miyachi
- Department of Physical Therapy, Faculty of Health Care Sciences, Hokuriku University, Kanazawa, Ishikawa 920-1154, Japan
| | - Kento Sakane
- Department of Physical Therapy, Faculty of Health Sciences, Kyoto Tachibana University, Kyoto 607-8175, Japan
| | - Saki Sugimoto
- Department of Physical Therapy, Faculty of Health Sciences, Kyoto Tachibana University, Kyoto 607-8175, Japan
| | - Airi Yamaguchi
- Department of Physical Therapy, Faculty of Health Sciences, Kyoto Tachibana University, Kyoto 607-8175, Japan
| | - Koki Yamamoto
- Department of Physical Therapy, Faculty of Health Sciences, Kyoto Tachibana University, Kyoto 607-8175, Japan
| |
Collapse
|
2
|
Wright AL, Weible AP, Estes OB, Wehr M. Ketamine does not rescue plaque load or gap detection in the 5XFAD mouse model of Alzheimer's disease. Front Aging Neurosci 2025; 17:1505908. [PMID: 39963471 PMCID: PMC11830726 DOI: 10.3389/fnagi.2025.1505908] [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: 10/04/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Ketamine has received growing attention for its effects on neuroplasticity and neuroinflammation, and as a treatment for depression and other mental health disorders. Recent evidence suggests that early sensory and behavioral deficits in Alzheimer's disease could be caused by synaptic disruption that occurs before irreversible neuropathology. This raises the possibility that ketamine could slow down or prevent network disruption and the ensuing sensory and behavioral deficits in Alzheimer's. Here we tested this idea in the 5XFAD mouse model of Alzheimer's, using either an acute single injection of ketamine, or chronic daily injections over 15 weeks. We tested the effects of ketamine on both amyloid plaque load and on a behavioral auditory gap detection task that is an early Alzheimer's biomarker in both mice and humans. We found that ketamine had no effect on plaque load, nor any effect on gap detection, for either acute or chronic dosing. Chronic ketamine facilitated startle responses specifically in 5XFAD mice, but this could simply be related to experience-dependent effects on stress or habituation rather than any rescue effect of ketamine on Alzheimer's-related deficits. We did find robust correlations between gap detection deficits and plaque load in auditory cortex and in the caudal pontine reticular nucleus, demonstrating that the behavioral deficits seen in 5XFAD mice are directly related to amyloid accumulation in these brain regions, and confirming the validity of gap detection as an early biomarker of Alzheimer's. Ketamine, however, had no effect on the strength of these correlations. We conclude that ketamine has no beneficial effect on the development of behavioral gap detection deficits or plaque load in the 5XFAD Alzheimer's mouse model, following either an acute single dose or a chronic daily dose regimen.
Collapse
Affiliation(s)
| | | | | | - Michael Wehr
- Department of Psychology, Institute of Neuroscience, University of Oregon, Eugene, OR, United States
| |
Collapse
|
3
|
Talaee N, Azad Yekta M, Vaseghi S. New insights into individual differences in response to chronic unpredictable mild stress (CUMS) in rats with respect to hippocampal BDNF and GSK3-β expression levels. Physiol Behav 2024; 287:114718. [PMID: 39426694 DOI: 10.1016/j.physbeh.2024.114718] [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: 09/11/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
Preclinical and clinical studies have shown a wide-range of individual differences in response to stressors or novel environments which can affect the susceptibility to develop abnormal behaviors and neuropsychiatric disorders. Both vulnerability and resiliency have been observed in animals and humans experiencing stressful events. Chronic unpredictable mild stress (CUMS) is a rodent depression model consisting of various stressors. This protocol leads to depressive- and anhedonic-like behaviors in rodents. The present study aimed to evaluate potential individual differences in response to CUMS in rats, with respect to the expression level of brain-derived neurotrophic factor (BDNF) and glycogen synthase kinases 3-beta (GSK3-β) (proteins involved in the modulation of mood, neuroplasticity, and cognition) in the hippocampus. CUMS was performed for four consecutive weeks. Depressive-like behavior, locomotor activity, anxiety-like behavior, and pain threshold were also evaluated using forced swim test (FST), open field test (OFT), and the hot plate (HP), respectively. Real-time PCR was used to evaluate BDNF and GSK3-β expression levels. The results showed that CUMS rats can be classified as two clusters: affected and non-affected (depressed and non-depressed). Affected rats showed depressive- and anxiety-like behaviors, decreased locomotor activity, and increased pain threshold. However, non-affected rats were similar to controls. In addition, there was a downregulation of BDNF and upregulation of GSK3-β in affected rats. Spearman correlation analysis also showed a relationship between BDNF and GSK3-β expression levels with individual differences. In conclusion, the present study showed that BDNF and GSK3-β may be involved in individual differences in CUMS rats.
Collapse
Affiliation(s)
- Nastaran Talaee
- Department of Psychology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mehrnaz Azad Yekta
- Department of Psychology, Faculty of Educational Sciences and Psychology, Islamshahr Branch, Islamic Azad University, Islamshahr, Iran.
| | - Salar Vaseghi
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran; Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| |
Collapse
|
4
|
Wang L, Zhao S, Shao J, Su C. The effect and mechanism of low-dose esketamine in neuropathic pain-related depression-like behavior in rats. Brain Res 2024; 1843:149117. [PMID: 38977235 DOI: 10.1016/j.brainres.2024.149117] [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: 01/09/2024] [Revised: 05/28/2024] [Accepted: 07/05/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND Clinical evidence suggests that Esketamine (ESK) is an effective treatment for depression. However, the effects of Esketamine in treating depression-like behavior induced by neuropathic pain is unclear. The underlying molecular mechanisms require further investigation to provide new therapeutic targets for the treatment of clinical neuropathic pain-related depression. METHODS A neuropathic pain-related depression model was established in rats with spared nerve injury (SNI). Male Sprague-Dawley rats were randomly divided into four groups: Sham Group, SNI group, SNI + Normal Saline (NS) Group and SNI + ESK5mg/kg Group. Mechanical pain thresholds were measured to assess pain sensitivity in SNI rats. On the 14th day after surgery a forced swim test and sucrose preference test were used to evaluate the depressive-like behavior of rats in each group. Further, a proteomic analysis was used to quantify differentially expressed proteins. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were analyzed to explore the main protein targets of SNI in the medial prefrontal cortex. The expression of proteins was detected by Western blotting. RESULTS A neuropathic pain-related depression model was established. Compared with the Sham group, the mechanical pain threshold was decreased significantly (13.2 ± 1.0 vs. 0.7 ± 0.01 g n = 8), while immobility on the forced swim test was also decreased (93.1 ± 7.4 vs. 169.5 ± 9.6 s n = 8), and sucrose preference rate was significantly increased (98.8 ± 0.3 vs. 73.1 ± 1.4n = 7) in SNI group rats. Compared with the SNI + NS group, the mechanical pain threshold was not statistically significant, while immobility on the forced swim test was clearly decreased (161.1 ± 11.6 vs. 77.9 ± 5.0 s n = 8), and sucrose preference rate was significantly increased (53.1 ± 8.9 vs. 96.1 ± 1.4n = 7) in SNI + ESK5mg/kg group rats. To further investigate the underlying mechanism, we employed proteomics to identify proteins exhibiting more than a 1.2-fold difference (P < 0.05) in expression levels within each group for subsequent analysis. Relative to the Sham group, 88 downregulated and 104 up-regulated proteins were identified in the SNI group, while 120 and 84 proteins were up- and down-regulated in the Esketamine treatment group compared with the SNI + NS group. Compared with Sham group, the expressions of mGluR5 and Homer1a were up-regulated in the medial prefrontal cortex (mPFC) in SNI group (mGluR5:0.97 ± 0.05 vs 1.47 ± 0.15, Homer1a:1.03 ± 0.06 vs 1.46 ± 0.16n = 6), and down-regulated after intervention with Esketamine (mGluR5:1.54 ± 0.11 vs 1.06 ± 0.07, Homer1a:1.51 ± 0.13 vs 1.12 ± 0.34n = 6). CONCLUSIONS Low-dose Esketamine appeared to relieve depression-like behavior induced by neuropathic pain. The Homer1a-mGluR5 signaling pathway might be the mechanism of antidepressant effect of Esketamine.
Collapse
Affiliation(s)
- Lijuan Wang
- Department of Anesthesiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha, Hunan, China; Department of Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Shuwu Zhao
- Department of Anesthesiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha, Hunan, China
| | - Jiali Shao
- Department of Anesthesiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha, Hunan, China
| | - Chen Su
- Department of Anesthesiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha, Hunan, China.
| |
Collapse
|
5
|
O'Brien JA, Austin PJ. Minocycline Abrogates Individual Differences in Nerve Injury-Evoked Affective Disturbances in Male Rats and Prevents Associated Supraspinal Neuroinflammation. J Neuroimmune Pharmacol 2024; 19:30. [PMID: 38878098 PMCID: PMC11180027 DOI: 10.1007/s11481-024-10132-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 06/08/2024] [Indexed: 06/19/2024]
Abstract
Chronic neuropathic pain precipitates a complex range of affective and behavioural disturbances that differ markedly between individuals. While the reasons for differences in pain-related disability are not well understood, supraspinal neuroimmune interactions are implicated. Minocycline has antidepressant effects in humans and attenuates affective disturbances in rodent models of pain, and acts by reducing neuroinflammation in both the spinal cord and brain. Previous studies, however, tend not to investigate how minocycline modulates individual affective responses to nerve injury, or rely on non-naturalistic behavioural paradigms that fail to capture the complexity of rodent behaviour. We investigated the development and resolution of pain-related affective disturbances in nerve-injured male rats by measuring multiple spontaneous ethological endpoints on a longitudinal naturalistic foraging paradigm, and the effect of chronic oral minocycline administration on these changes. Disrupted foraging behaviours appeared in 22% of nerve-injured rats - termed 'affected' rats - and were present at day 14 but partially resolved by day 21 post-injury. Minocycline completely prevented the emergence of an affected subgroup while only partly attenuating mechanical allodynia, dissociating the relationship between pain and affect. This was associated with a lasting downregulation of ΔFosB expression in ventral hippocampal neurons at day 21 post-injury. Markers of microglia-mediated neuroinflammation were not present by day 21, however proinflammatory microglial polarisation was apparent in the medial prefrontal cortex of affected rats and not in CCI minocycline rats. Individual differences in affective disturbances following nerve injury are therefore temporally related to altered microglial morphology and hippocampal neuronal activation, and are abrogated by minocycline.
Collapse
Affiliation(s)
- Jayden A O'Brien
- Brain and Mind Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Paul J Austin
- Brain and Mind Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
6
|
Hashimoto K. Are "mystical experiences" essential for antidepressant actions of ketamine and the classic psychedelics? Eur Arch Psychiatry Clin Neurosci 2024:10.1007/s00406-024-01770-7. [PMID: 38411629 DOI: 10.1007/s00406-024-01770-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/22/2024] [Indexed: 02/28/2024]
Abstract
The growing interest in the rapid and sustained antidepressant effects of the dissociative anesthetic ketamine and classic psychedelics, such as psilocybin, is remarkable. However, both ketamine and psychedelics are known to induce acute mystical experiences; ketamine can cause dissociative symptoms such as out-of-body experience, while psychedelics typically bring about hallucinogenic experiences, like a profound sense of unity with the universe or nature. The role of these mystical experiences in enhancing the antidepressant outcomes for patients with depression is currently an area of ongoing investigation and debate. Clinical studies have shown that the dissociative symptoms following the administration of ketamine or (S)-ketamine (esketamine) are not directly linked to their antidepressant properties. In contrast, the antidepressant potential of (R)-ketamine (arketamine), thought to lack dissociative side effects, has yet to be conclusively proven in large-scale clinical trials. Moreover, although the activation of the serotonin 5-HT2A receptor is crucial for the hallucinogenic effects of psychedelics in humans, its precise role in their antidepressant action is still under discussion. This article explores the importance of mystical experiences in enhancing the antidepressant efficacy of both ketamine and classic psychedelics.
Collapse
Affiliation(s)
- Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, 1-8-1 Inohana, Chiba, 260-8670, Japan.
| |
Collapse
|
7
|
Wu GH, Guo QH, Xu XD, Lin JC, You GT, Lin CH, Zhang LC. Ketamine exerts dual effects on the apoptosis of primary cultured hippocampal neurons from fetal rats in vitro. Metab Brain Dis 2023; 38:2417-2426. [PMID: 37273081 DOI: 10.1007/s11011-023-01236-0] [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: 09/06/2022] [Accepted: 05/16/2023] [Indexed: 06/06/2023]
Abstract
Ketamine, a noncompetitive N-methyl D-aspartate (NMDA) receptor antagonist, is widely used in pediatric clinical practice. The neuroprotective and neurotoxic effects of ketamine on brain neurons during development remain controversial. The reason may be related to the different concentrations of ketamine used in practice and the small range of concentrations used in previous studies. In this study, cultured hippocampal neurons were treated with ketamine in a wide range of concentrations to comprehensively observe the effects of different concentrations of ketamine on neurons. We demonstrated that low concentrations of ketamine (10 μM, 100 μM and 1000 μM) promoted neuronal survival (p < 0.05) and reduced neuronal apoptosis (p < 0.05) compared with those of the control group. High concentrations of ketamine (2000 μM, 2500 μM and 3000 μM) reduced neuronal survival (p < 0.05) and promoted neuronal apoptosis (p < 0.05). The p38 MAPK inhibitor SB203580 reduced neuronal apoptosis induced by high concentrations of ketamine (2500 μM) (p < 0.05). Our findings indicate that ketamine exerts a dual effect on the apoptosis of primary cultured fetal rat hippocampal neurons in vitro and that the neurotoxic effects of ketamine are related to activation of the p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Guo-Hua Wu
- Fujian Provincial Key Laboratory of Burn and Trauma, Fujian Burn Institute, Fujian Burn Medical Center, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Que-Hui Guo
- Department of Ultrasound, the Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, 350003, Fujian, China
| | - Xiao-Dong Xu
- Department of Anesthesiology, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fujian, 350001, Fuzhou, China
| | - Jian-Chang Lin
- Fujian Provincial Key Laboratory of Burn and Trauma, Fujian Burn Institute, Fujian Burn Medical Center, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Gui-Ting You
- Department of Neurosurgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Cai-Hou Lin
- Department of Neurosurgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China.
| | - Liang-Cheng Zhang
- Department of Anesthesiology, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fujian, 350001, Fuzhou, China.
| |
Collapse
|
8
|
Tao W, Su K, Huang Y, Lu Z, Wang Y, Yang L, Zhang G, Liu W. Zuojinwan ameliorates CUMS-induced depressive-like behavior through inducing ubiquitination of MyD88 via SPOP/MyD88/NF-κB pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 312:116487. [PMID: 37059253 DOI: 10.1016/j.jep.2023.116487] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/24/2023] [Accepted: 04/09/2023] [Indexed: 05/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zuojinwan (ZJW) is a traditional Chinese medicine compound, which is often used clinically to treat gastritis and has anti-inflammatory activity. It was found that ZJW is involved in suppressing the expression of inflammatory factors, and neuroinflammation is thought to be associated with the development of depression. AIM OF THE STUDY In this study, we investigated whether ZJW could exert antidepressant effects by regulating MyD88 ubiquitination in depressed mice and attempted to elucidate the possible mechanisms. MATERIALS AND METHODS Six active compounds of Zuojinwan (ZJW) were identified by HPLC. Then, the effects of ZJW on depression-like behavior in mice were investigated by constructing a chronic unpredictable mild stimulation (CUMS) mouse model. Meanwhile, the effect of ZJW on hippocampal neurons was investigated by Nissl staining. In addition, western blotting, PCR, ELISA, co-immunoprecipitation and immunostaining were used to explore whether ZJW could inhibit neuroinflammation through SPOP/MyD88/NF-κB pathway and thus produce antidepressant effects. Finally, we constructed the AAV-Sh-SPOP virus vector to silence SPOP and verify the mechanism of ZJW's antidepressant action. RESULTS ZJW could dramatically ameliorate the depressive behavior induced by CUMS stimulation and alleviate hippocampal neuronal damage. CUMS stimulation resulted in decreased SPOP expression, impaired MyD88 ubiquitination, and activation of downstream NF-κB signaling, which could be reversed by ZJW. In addition, ZJW could significantly ameliorate the abnormal activation of microglia, and the excessive levels of pro-inflammatory factors were inhibited. By blocking the expression of SPOP, we found that ZJW exerted anti-inflammatory and antidepressant effects mainly by promoting the ubiquitination of MyD88 and inhibiting the activation of downstream inflammatory signals. CONCLUSION In conclusion, ZJW possesses alleviating effects on depression induced by CUMS stimulation. ZJW can inhibit neuroinflammation and improve neuroinflammation-induced depression-like behaviors through SPOP/MyD88/NF-κB pathway.
Collapse
Affiliation(s)
- Weiwei Tao
- Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China; School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Kunhan Su
- Department of Gastroenterology, Nanjing Integrated Traditional Chinese and Western Medicine Hospital, Nanjing, 210014, China
| | - Yuzhen Huang
- Department of Gastroenterology, Nanjing Integrated Traditional Chinese and Western Medicine Hospital, Nanjing, 210014, China
| | - Zihan Lu
- China Pharmaceutical University, Nanjing, 210009, China
| | - Yan Wang
- Department of Gastroenterology, Nanjing Integrated Traditional Chinese and Western Medicine Hospital, Nanjing, 210014, China; Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lu Yang
- Department of Gastroenterology, Nanjing Integrated Traditional Chinese and Western Medicine Hospital, Nanjing, 210014, China; Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Guoying Zhang
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wanli Liu
- Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China; Department of Gastroenterology, Nanjing Integrated Traditional Chinese and Western Medicine Hospital, Nanjing, 210014, China.
| |
Collapse
|
9
|
Lullau APM, Haga EMW, Ronold EH, Dwyer GE. Antidepressant mechanisms of ketamine: a review of actions with relevance to treatment-resistance and neuroprogression. Front Neurosci 2023; 17:1223145. [PMID: 37614344 PMCID: PMC10442706 DOI: 10.3389/fnins.2023.1223145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/12/2023] [Indexed: 08/25/2023] Open
Abstract
Concurrent with recent insights into the neuroprogressive nature of depression, ketamine shows promise in interfering with several neuroprogressive factors, and has been suggested to reverse neuropathological patterns seen in depression. These insights come at a time of great need for novel approaches, as prevalence is rising and current treatment options remain inadequate for a large number of people. The rapidly growing literature on ketamine's antidepressant potential has yielded multiple proposed mechanisms of action, many of which have implications for recently elucidated aspects of depressive pathology. This review aims to provide the reader with an understanding of neuroprogressive aspects of depressive pathology and how ketamine is suggested to act on it. Literature was identified through PubMed and Google Scholar, and the reference lists of retrieved articles. When reviewing the evidence of depressive pathology, a picture emerges of four elements interacting with each other to facilitate progressive worsening, namely stress, inflammation, neurotoxicity and neurodegeneration. Ketamine acts on all of these levels of pathology, with rapid and potent reductions of depressive symptoms. Converging evidence suggests that ketamine works to increase stress resilience and reverse stress-induced dysfunction, modulate systemic inflammation and neuroinflammation, attenuate neurotoxic processes and glial dysfunction, and facilitate synaptogenesis rather than neurodegeneration. Still, much remains to be revealed about ketamine's antidepressant mechanisms of action, and research is lacking on the durability of effect. The findings discussed herein calls for more longitudinal approaches when determining efficacy and its relation to neuroprogressive factors, and could provide relevant considerations for clinical implementation.
Collapse
Affiliation(s)
- August P. M. Lullau
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Emily M. W. Haga
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Eivind H. Ronold
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Gerard E. Dwyer
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
- NORMENT Centre of Excellence, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
10
|
VanderZwaag J, Halvorson T, Dolhan K, Šimončičová E, Ben-Azu B, Tremblay MÈ. The Missing Piece? A Case for Microglia's Prominent Role in the Therapeutic Action of Anesthetics, Ketamine, and Psychedelics. Neurochem Res 2023; 48:1129-1166. [PMID: 36327017 DOI: 10.1007/s11064-022-03772-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 08/25/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
There is much excitement surrounding recent research of promising, mechanistically novel psychotherapeutics - psychedelic, anesthetic, and dissociative agents - as they have demonstrated surprising efficacy in treating central nervous system (CNS) disorders, such as mood disorders and addiction. However, the mechanisms by which these drugs provide such profound psychological benefits are still to be fully elucidated. Microglia, the CNS's resident innate immune cells, are emerging as a cellular target for psychiatric disorders because of their critical role in regulating neuroplasticity and the inflammatory environment of the brain. The following paper is a review of recent literature surrounding these neuropharmacological therapies and their demonstrated or hypothesized interactions with microglia. Through investigating the mechanism of action of psychedelics, such as psilocybin and lysergic acid diethylamide, ketamine, and propofol, we demonstrate a largely under-investigated role for microglia in much of the emerging research surrounding these pharmacological agents. Among others, we detail sigma-1 receptors, serotonergic and γ-aminobutyric acid signalling, and tryptophan metabolism as pathways through which these agents modulate microglial phagocytic activity and inflammatory mediator release, inducing their therapeutic effects. The current review includes a discussion on future directions in the field of microglial pharmacology and covers bidirectional implications of microglia and these novel pharmacological agents in aging and age-related disease, glial cell heterogeneity, and state-of-the-art methodologies in microglial research.
Collapse
Affiliation(s)
- Jared VanderZwaag
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Torin Halvorson
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Kira Dolhan
- Department of Psychology, University of Victoria, Vancouver, BC, Canada
- Department of Biology, University of Victoria, Vancouver, BC, Canada
| | - Eva Šimončičová
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Benneth Ben-Azu
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Marie-Ève Tremblay
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Département de médecine moléculaire, Université Laval, Québec City, QC, Canada.
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada.
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
- Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
11
|
Wufuer D, Aierken H, Liang Z, Zheng JP, Li L. Association between comorbid asthma and depression and depression-related gene SNPs. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2023. [DOI: 10.1016/j.jrras.2022.100496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
12
|
Antidepressant Effect of Ketamine on Inflammation-Mediated Cytokine Dysregulation in Adults with Treatment-Resistant Depression: Rapid Systematic Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1061274. [PMID: 36160713 PMCID: PMC9507757 DOI: 10.1155/2022/1061274] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/25/2022] [Accepted: 08/23/2022] [Indexed: 11/28/2022]
Abstract
Background Major depressive disorder (MDD) and treatment-resistant depression (TRD) represent a global source of societal and health burden. To advise proper management of inflammation-related depression among TRD patients, it is important to identify therapeutic clinical treatments. A key factor is related to proinflammatory cytokines such as interleukin- (IL-) 1β, IL-6, and tumor necrosis factor- (TNF-) α which have been implicated in the pathogenesis of depressive symptoms in MDD patients. Ketamine may provide an anti-inflammatory therapeutic strategy by targeting proinflammatory pathways associated with depressive disorders, which may be exacerbated in the ageing population with TRD. Objective Despite a burgeoning body of literature demonstrating that inflammation is linked to TRD, there is still a lack of comprehensive research on the relationship between proinflammatory biomarkers and ketamine's antidepressant effect on TRD patients. Method The Cochrane Library and PubMed/MEDLINE databases were systematically searched from inception up to February 1, 2022, adopting broad inclusion criteria to assess clinical topics related to the impact of ketamine on inflammatory cytokines in TRD patients. The present work is in compliance with the World Health Organization Rapid Review Guide. Results Five out of the seven studies examined in this review show that ketamine infusion may reduce depressive symptoms with a quick start of effect on TRD patients. Based on the Montgomery-Åsberg Depression Rating Scale (MADRS) and Hamilton Depression Rating Scale (HAM-D) scores, the overall response rate for ketamine was 56%; that is, 56% of those treated with ketamine had MADRS/HAM-D scores decreased by at least 50%. Conclusions While the anti-inflammatory effects of ketamine modulate specific proinflammatory cytokines, its rapid antidepressant effect on TRD patients remains inconsistent. However, our study findings can provide a reliable basis for future research on how to improve systemic inflammatory immune disorders and mental health. We suggest that ketamine infusion may be part of a comprehensive treatment approach in TRD patients with elevated levels of depression-specific inflammatory biomarkers.
Collapse
|
13
|
A Green Approach for the Biosynthesis of Gold Nanoparticles Using Cuminum cyminum L. Seed and Its Application for Pain Management in Rats. IRANIAN BIOMEDICAL JOURNAL 2022; 26:219-29. [PMID: 35280043 PMCID: PMC9440691 DOI: 10.52547/ibj.26.3.219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
14
|
Ren J, Yu L, Lin J, Ma L, Gao DS, Sun N, Liu Y, Fang L, Cheng Z, Sun K, Yan M. Dimethyl itaconate inhibits neuroinflammation to alleviate chronic pain in mice. Neurochem Int 2022; 154:105296. [PMID: 35121012 DOI: 10.1016/j.neuint.2022.105296] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/21/2022] [Accepted: 01/29/2022] [Indexed: 10/19/2022]
Abstract
The metabolite itaconate has both anti-inflammatory and immunomodulatory effects. However, its influence on chronic pain is unclear. Here, we demonstrated that intraperitoneal injection of the itaconate derivative dimethyl itaconate (DI) alleviates chronic pain symptoms, such as allodynia and hyperalgesia, in spinal nerve ligation (SNL) and inflammatory pain models. Moreover, intraperitoneal DI reduced the secretion of inflammatory cytokines (i.e., interleukin-1β, tumour necrosis factor-alpha) in dorsal root ganglion (DRG), spinal cord and hind paw tissues, suppressed the activation of macrophages in DRG and glial cells in the spinal dorsal horn and decreased the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) in the DRG and spinal cord. DI boosted nuclear factor-erythroid 2 p45-related factor 2 (Nrf2) levels in the DRG and spinal cord of SNL mice. Intraperitoneal administration of the Nrf2 inhibitor ML385 abolished the analgesic effect of DI and decreased the expression of Nrf2 in the DRG and spinal cord. Similarly, administration of DI potently reversed the lipopolysaccharide (LPS)-induced inflammatory effect in microglia. Reduction of endogenous itaconate levels by pretreatment with immune-responsive gene 1 (IRG1) siRNA blocked Nrf2 expression, which impaired the analgesic and anti-inflammatory effects of DI in vitro. Therefore, our findings reveal for the first time that intraperitoneal DI elicits anti-inflammatory effect and sustained chronic pain relief, which may be regarded as a promising therapeutic agent for chronic pain treatment.
Collapse
Affiliation(s)
- Jinxuan Ren
- Department of Anesthesiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lina Yu
- Department of Anesthesiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiaqi Lin
- Department of Anesthesiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Longfei Ma
- Department of Anesthesiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Dave Schwinn Gao
- Department of Anesthesiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Na Sun
- Department of Anesthesiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ying Liu
- Department of Anesthesiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lili Fang
- Department of Anesthesiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhenzhen Cheng
- Department of Anesthesiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kai Sun
- Department of Anesthesiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Min Yan
- Department of Anesthesiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
15
|
Best SRD, Haustrup N, Pavel DG. Brain SPECT as an Imaging Biomarker for Evaluating Effects of Novel Treatments in Psychiatry-A Case Series. Front Psychiatry 2022; 12:713141. [PMID: 35095582 PMCID: PMC8793864 DOI: 10.3389/fpsyt.2021.713141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 12/13/2021] [Indexed: 01/23/2023] Open
Abstract
The difficulties of evaluating patients with complex neuropsychiatric conditions and prescribing appropriate treatments are well known. Imaging complements clinical assessments and allows a clinician to narrow the differential diagnosis by facilitating accurate and efficient evaluation. This is particularly relevant to neuropsychiatric conditions that are often diagnosed using a trial-and error process of exclusion. Single Photon Emission Computed Tomography (SPECT) is a functional brain imaging procedure that allows practitioners to measure the functional changes of gray matter structures based on regional cerebral blood flow (rCBF). The accurate diagnosis and treatment selection in psychiatry is challenging due to complex cases and frequent comorbidities. However, such complex neuropsychiatric conditions are increasingly benefitting from new treatment approaches, in addition to established medications. Among these are combination transcranial magnetic stimulation with ketamine infusions (CTK), hyperbaric oxygen therapy (HBOT) and perispinal administration of etanercept (PSE). This article provides readers with six case study examples that demonstrate how brain SPECT imaging can be used, both as a diagnostic tool, and as a potential biomarker for monitoring and evaluating novel treatments for patients with complex neuropsychiatric conditions. Six patients were assessed in our clinic and baseline brain SPECT imagesTourettes and a long history of alcohol were visually compared with SPECT images collected after periods of treatment with CTK or HBOT followed by PSE. This retrospective review demonstrates the clinical utility of these novel treatments and describes how SPECT imaging can complement standard diagnostic assessments. A novel display technique for SPECT images is described and we argue that SPECT imaging can be used for monitoring biomarker for clinical change.
Collapse
Affiliation(s)
| | | | - Dan G. Pavel
- PathFinder Brain SPECT, Deerfield, IL, United States
| |
Collapse
|
16
|
Ren H, Han R, Liu X, Wang L, Koehler RC, Wang J. Nrf2-BDNF-TrkB pathway contributes to cortical hemorrhage-induced depression, but not sex differences. J Cereb Blood Flow Metab 2021; 41:3288-3301. [PMID: 34238051 PMCID: PMC8669278 DOI: 10.1177/0271678x211029060] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Post-stroke depression, observed in 30-50% of stroke patients, negatively affects quality of life and mortality. The pathogenesis of post-stroke depression is complex, but heightened reactive oxygen species production and inflammation might be two key factors. We have reported that intracerebral hemorrhage (ICH) in cerebral cortex produces depression-like behavior in young male mice. Here, we found that mice lacking nuclear factor erythroid-derived 2-related factor 2 (Nrf2), a transcription factor that upregulates antioxidant proteins and trophic factors such as brain-derived neurotrophic factor (BDNF), had more severe depression-like behavior than wild-type mice at days 21 to 28 after cortical ICH (c-ICH). Moreover, the expression of Nrf2, heme oxygenase-1, BDNF, and TrkB were significantly decreased in wild-type mice after c-ICH. Interestingly, TP-500 (2 mg/kg), a potent Nrf2 inducer, decreased the inflammatory response and reactive oxygen species production on day 28 after c-ICH and improved depression-like behaviors. TrkB receptor antagonist ANA-12 abolished this anti-depression effect. Depression was more severe in female than in male wild-type mice after ICH, but TP-500 improved depression-like behavior in females. These results suggest that downregulation of Nrf2-BDNF-TrkB signaling contributes to development of post-stroke depression, and that Nrf2 inducer TP-500 might improve depression after c-ICH.
Collapse
Affiliation(s)
- Honglei Ren
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University, Baltimore, MD, USA
| | - Ranran Han
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University, Baltimore, MD, USA
| | - Xi Liu
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University, Baltimore, MD, USA
| | - Limin Wang
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University, Baltimore, MD, USA
| | - Raymond C Koehler
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University, Baltimore, MD, USA
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
17
|
Zhao Z, Xu Q, Chen Y, Liu C, Zhang F, Han Y, Cao J. The effect of low-dose ketamine on postoperative quality of recovery in patients undergoing breast cancer surgery: A randomised, placebo-controlled trial. Int J Clin Pract 2021; 75:e15010. [PMID: 34807494 DOI: 10.1111/ijcp.15010] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/24/2021] [Accepted: 11/19/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Low-dose ketamine has been proved to reduce opioid consumption, prevent depressant action and improve postoperative analgesia. Women undergoing mastectomy experience may not only have persistent postoperative pain syndromes but also emotional problems. However, the effect of intraoperative infusion of low-dose ketamine on postoperative quality of recovery among these patients has not yet been fully studied. METHODS In this prospective, randomised, single-centre trial, 100 patients planned for modified radical mastectomy were randomly assigned to one of two groups: control group (group C) or ketamine group (group K) at a ratio of 1:1. Group K received the bolus dose of 0.5 mg/kg ketamine and followed by 0.25 mg·kg-1 ·h-1 after the compliment of anaesthesia induction until the end of the surgery, whilst group C received an equivalent dose and regiment of normal saline was group K. The primary outcome was to assess the effects of low-dose ketamine on postoperative quality of recovery using the 40-Item Quality of Recovery (QoR-40) scale on a postoperative day 1 (POD1). The secondary outcome was to assess the numeric rating scale (NRS) at 4, 24 and 48 h after the operation, identity-consequence fatigue scale (ICFS) scores at 3 and 7 days after the operation, hospital anxiety and depression scale (HADS) scores at 2 days and 3 months, as well as chronic pain at 3 months. In a post hoc analysis, the 5 subsections of the QoR-40 scores were also analysed. RESULTS A total of 100 subjects were randomised. The primary outcome of QoR-40 scores on POD1 was available in 97 patients (49 in group C and 48 in group K). Global QoR-40 scores were not significantly different between group C and group K (169.8 ± 10.7 vs. 172.7 ± 7.5, 95% CI -1.35 (-5.50, 2.80), p = .519). In a post hoc analysis, pain scores were significantly higher in group K than in group C (29.8 ± 3.8 vs. 31.7 ± 2.1, 95% CI -1.81 (-3.00, -0.62), p = .003). The secondary outcomes, including NRS, ICFS scores, HADS scores and chronic pain had no difference between groups (p value for each >.15). CONCLUSION Intraoperative low-dose ketamine infusion did not improve the overall quality of recovery on POD 1 in patients undergoing breast cancer surgery.
Collapse
Affiliation(s)
- Zijian Zhao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou City, China
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing City, China
| | - Qiqi Xu
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing City, China
- Department of Anesthesiology, Huai'an First People's Hospital, Huai'an City, China
| | - Yao Chen
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou City, China
| | - Chen Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou City, China
| | - Fangfang Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou City, China
| | - Yuan Han
- Department of Anesthesiology, Eye & ENT Hospital of Fudan University, Shanghai City, China
| | - Junli Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou City, China
| |
Collapse
|
18
|
Zhang L, Zhang H, Xie J, Wang X. Identification of Gene Co-Expression Modules and Core Genes Related to Immune Disorders in Major Depression Disorder. Int J Gen Med 2021; 14:7983-7993. [PMID: 34785941 PMCID: PMC8591119 DOI: 10.2147/ijgm.s336686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/26/2021] [Indexed: 12/16/2022] Open
Abstract
Introduction Various studies have confirmed the connection between the mental state and the immune system, that is, mental activities can regulate immune function, and immune system disorders can not only lead to bodily diseases but also changes related to mentality, behavior, personality, and aging. However, the specific regulatory mechanism and key genes are still unclear. Methods We obtained the peripheral blood gene sequencing data from patients with major depression and normal volunteers from the GEO database and evaluated the scores of different immune cells by immune scoring algorithm. Using the immune scores as clinical data, a weighted gene co-expression network analysis (WGCNA) was carried out to study the association between the clinical characteristics and modules. Therefore, providing an opportunity to lock modules and core genes which are highly related to the immune regulation of major depression. Results Thirteen co-expression modules were clustered from 20,011 genes, the yellow module had a positive correlation with CD4+ T cell, CD8+ T cell, B cell, and NK cell immune scores, and a negative correlation with purple module. Functional annotation and signaling pathway analysis illustrated that the yellow module is mostly enriched in thymus development, T cell co-stimulation and differentiation, and B cell activation. Genes in the purple module were primarily related to inhibition of protein phosphorylation, leukocyte migration, promotion of apoptosis and hypoxia and other signaling pathways. Additionally, hub genes in the yellow and purple modules were detected, in which SKAP1 and RALB may be important regulatory genes affecting the immune status of patients with depression. Discussion In general, our study reveals the key genes related to the decrease in CD4+ T cells, CD8+ T cells, and B cells, in the peripheral blood of patients with depression, which provides some new insights and understandings for the clinical treatment and diagnosis of major depression. Drug design targeting these targets may provide the possibility for the treatment of major depression.
Collapse
Affiliation(s)
- Lei Zhang
- School of Public Administration, Hohai University, Nanjing, People's Republic of China
| | - Haibo Zhang
- Organization of Personnel Division, Jiangsu Provincial People's Hospital (The First Affiliated Hospital of Nanjing Medical University), Nanjing, People's Republic of China
| | - Jiadong Xie
- School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Xu Wang
- Xuzhou Medical University, Xuzhou, People's Republic of China
| |
Collapse
|
19
|
Role of neuroglia in neuropathic pain and depression. Pharmacol Res 2021; 174:105957. [PMID: 34688904 DOI: 10.1016/j.phrs.2021.105957] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/06/2021] [Accepted: 10/20/2021] [Indexed: 12/30/2022]
Abstract
Patients with neuropathic pain induced by nerve injury usually present with co-morbid affective changes, such as depression. Neuroglia was reported to play an important role in the development and maintenance of neuropathic pain both centrally and peripherally. Meanwhile, there have been studies showing that neuroglia participated in the development of depression. However, the specific role of neuroglia in neuropathic pain and depression has not been reviewed comprehensively. Therefore, we summarized the recent findings on the role of neuroglia in neuropathic pain and depression. Based on this review, we found a bridge-like role of neuroglia in neuropathic pain co-morbid with depression. This review may provide therapeutic implications in the treatment of neuropathic pain and offer potential help in the studies of mechanisms in the future.
Collapse
|
20
|
Plasma inflammatory cytokines and treatment-resistant depression with comorbid pain: improvement by ketamine. J Neuroinflammation 2021; 18:200. [PMID: 34526064 PMCID: PMC8444441 DOI: 10.1186/s12974-021-02245-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/22/2021] [Indexed: 12/13/2022] Open
Abstract
Background Treatment-resistant depression (TRD) and pain frequently coexist clinically. Ketamine has analgesic and antidepressant effects, but few studies have evaluated individual differences in antidepressant outcomes to repeated ketamine in TRD patients with comorbid pain. Our aims were to determine the difference in ketamine’s antidepressant effects in TRD patients with or without pain and then to examine whether inflammatory cytokines might contribute to ketamine’s effect. Methods Sixty-six patients with TRD received six infusions of ketamine. Plasma levels of 19 inflammatory cytokines were assessed at baseline and post-infusion (day 13 and day 26) using the Luminex assay. Plasma inflammatory cytokines of sixty healthy controls (HCs) were also examined. Results TRD patients with pain had a higher antidepressant response rate (χ2 = 4.062, P = 0.044) and remission rate (χ2 = 4.062, P = 0.044) than patients without pain. Before ketamine treatment, GM-CSF and IL-6 levels were higher in the pain group than in the non-pain and HC groups. In the pain group, levels of TNF-α and IL-6 at day 13 and GM-CSF, fractalkine, IFN-γ, IL-10, MIP-3α, IL-12P70, IL-17α, IL-1β, IL-2, IL-4, IL-23, IL-5, IL-6, IL-7, MIP-1β, and TNF-α at day 26 were lower than those at baseline; in the non-pain group, TNF-α levels at day 13 and day 26 were lower than those at baseline. In the pain group, the changes of IL-6 were associated with improvement in pain intensity (β = 0.333, P = 0.001) and depressive symptoms (β = 0.478, P = 0.005) at day 13. Path analysis showed the direct (β = 2.995, P = 0.028) and indirect (β = 0.867, P = 0.042) effects of changes of IL-6 on improvement in depressive symptoms both were statistically significant. Conclusion This study suggested that an elevated inflammatory response plays a critical role in individual differences in TRD patients with or without pain. Ketamine showed great antidepressant and analgesic effects in TRD patients with pain, which may be related to its effects on modulating inflammation. Trial registration ChiCTR, ChiCTR-OOC-17012239. Registered on 26 May 2017 Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02245-5.
Collapse
|
21
|
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: 49] [Impact Index Per Article: 12.3] [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
|
22
|
Wu Q, Wang B, Ntim M, Zhang X, Na XY, Yuan YH, Wu XF, Yang JY, Li S. SRC-1 Deficiency Increases Susceptibility of Mice to Depressive-Like Behavior After Exposure to CUMS. Neurochem Res 2021; 46:1830-1843. [PMID: 33881662 DOI: 10.1007/s11064-021-03316-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 11/24/2022]
Abstract
Steroid receptor coactivator 1 (SRC-1) is one of the coactivators recruited by the nuclear receptors (NRs) when NRs are activated by steroid hormones, such as glucocorticoid. SRC-1 is abundant in hippocampus and hypothalamus and is also related to some major risk factors for depression, implicated by its reduced expression after stress and its effect on hypothalamus-pituitary-adrenal gland axis function. However, whether SRC-1 is involved in the formation of depression remains unclear. In this study, we firstly established chronic unpredictable stress (CUS) to induce depressive-like behaviors in mice and found that SRC-1 expression was reduced by CUS. A large number of studies have shown that neuroinflammation is associated with stress-induced depression and lipopolysaccharide (LPS) injection can lead to neuroinflammation and depressive-like behaviors in mice. Our result indicated that LPS treatment also decreased SRC-1 expression in mouse brain, implying the involvement of SRC-1 in the process of inflammation and depression. Next, we showed that the chronic unpredictable mild stress (CUMS) failed to elicit the depressive-like behaviors and dramatically promoted the expression of SRC-1 in brain of wild type mice. What's more, the SRC-1 knockout mice were more susceptible to CUMS to develop depressive-like behaviors and presented the changed expression of glucocorticoid receptor. However, SRC-1 deficiency did not affect the microglia activation induced by CUMS. Altogether, these results indicate a correlation between SRC-1 level and depressive-like behaviors, suggesting that SRC-1 might be involved in the development of depression induced by stress.
Collapse
Affiliation(s)
- Qiong Wu
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Bin Wang
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Michael Ntim
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xuan Zhang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Xue-Yan Na
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Yu-Hui Yuan
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xue-Fei Wu
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China.
| | - Jin-Yi Yang
- Department of Urology, Affiliated Dalian Friendship Hospital of Dalian Medical University, Dalian, China.
| | - Shao Li
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China.
| |
Collapse
|
23
|
Zhang N, Yao L, Wang P, Liu Z. Immunoregulation and antidepressant effect of ketamine. Transl Neurosci 2021; 12:218-236. [PMID: 34079622 PMCID: PMC8155793 DOI: 10.1515/tnsci-2020-0167] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 04/04/2021] [Accepted: 04/06/2021] [Indexed: 12/15/2022] Open
Abstract
Major depressive disorder (MDD) is a common mental health disorder that brings severe disease burden worldwide. Traditional antidepressants are mainly targeted at monoamine neurotransmitters, with low remission rates and high recurrence rates. Ketamine is a noncompetitive glutamate N-methyl-d-aspartate receptor (NMDAR) antagonist, and its rapid and powerful antidepressant effects have come to light. Its antidepressant mechanism is still unclarified. Research found that ketamine had not only antagonistic effect on NMDAR but also strong immunomodulatory effect, both of which were closely related to the pathophysiology of MDD. Although there are many related studies, they are relatively heterogeneous. Therefore, this review mainly describes the immune mechanisms involved in MDD and how ketamine plays an antidepressant role by regulating peripheral and central immune system, including peripheral inflammatory cytokines, central microglia, and astrocytes. This review summarizes the related research, finds out the deficiencies of current research, and provides ideas for future research and the development of novel antidepressants.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Rd. 238, 430060, Wuhan, China
| | - Lihua Yao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Rd. 238, 430060, Wuhan, China
| | - Peilin Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Rd. 238, 430060, Wuhan, China
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Rd. 238, 430060, Wuhan, China
| |
Collapse
|
24
|
Down-regulation of MST1 in hippocampus protects against stress-induced depression-like behaviours and synaptic plasticity impairments. Brain Behav Immun 2021; 94:196-209. [PMID: 33607238 DOI: 10.1016/j.bbi.2021.02.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 01/26/2021] [Accepted: 02/05/2021] [Indexed: 12/15/2022] Open
Abstract
Depression is a common mental disorder, and its main environmental risk factor is chronic stress. The activation of mammalian STE20-like kinase 1 (MST1), a key factor involved in the underlying pathophysiology of stress, can trigger synaptic plasticity impairment, neuronal dysfunction and neuroinflammation. However, it is unclear whether down-regulation of MST1 in the hippocampus protects against stress-induced behavioural dysfunctions. In this study, three mouse models were used to assess the role of MST1 in stress. Various behavioural tests, in vivo electrophysiological recordings, Western blotting, Golgi staining and immunofluorescence assay were used. The data showed that the level of phospho-MST1 (T183) was significantly increased in the hippocampus of mice subjected to chronic unpredictable mild stress (CUMS) and that mice with MST1 overexpression showed depression-like behaviours. Importantly, the impairment of cognitive functions and the hippocampal synaptic plasticity induced by CUMS were significantly improved by MST1 knockdown, suggesting that MST1 down-regulation effectively protected against stress-induced behavioural dysfunctions. Moreover, MST1 knockdown suppressed CUMS-induced microglial activation, reduced the abnormal expression of inflammatory cytokines and impeded the activation of p38, implying that the antidepressant-like effects of MST1 knockdown were associated with inhibiting the p38 pathway. These findings suggest that hippocampal MST1 is an essential regulator of stress, which can be an ideal target for the development of antidepressants in the future.
Collapse
|
25
|
Warnecke A, Harre J, Staecker H, Prenzler N, Strunk D, Couillard‐Despres S, Romanelli P, Hollerweger J, Lassacher T, Auer D, Pachler K, Wietzorrek G, Köhl U, Lenarz T, Schallmoser K, Laner‐Plamberger S, Falk CS, Rohde E, Gimona M. Extracellular vesicles from human multipotent stromal cells protect against hearing loss after noise trauma in vivo. Clin Transl Med 2020; 10:e262. [PMID: 33377658 PMCID: PMC7752163 DOI: 10.1002/ctm2.262] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/03/2020] [Accepted: 12/06/2020] [Indexed: 12/21/2022] Open
Abstract
The lack of approved anti-inflammatory and neuroprotective therapies in otology has been acknowledged in the last decades and recent approaches are heralding a new era in the field. Extracellular vesicles (EVs) derived from human multipotent (mesenchymal) stromal cells (MSC) can be enriched in vesicular secretome fractions, which have been shown to exert effects (eg, neuroprotection and immunomodulation) of their parental cells. Hence, MSC-derived EVs may serve as novel drug candidates for several inner ear diseases. Here, we provide first evidence of a strong neuroprotective potential of human stromal cell-derived EVs on inner ear physiology. In vitro, MSC-EV preparations exerted immunomodulatory activity on T cells and microglial cells. Moreover, local application of MSC-EVs to the inner ear significantly attenuated hearing loss and protected auditory hair cells from noise-induced trauma in vivo. Thus, EVs derived from the vesicular secretome of human MSC may represent a next-generation biological drug that can exert protective therapeutic effects in a complex and nonregenerating organ like the inner ear.
Collapse
Affiliation(s)
- Athanasia Warnecke
- Department of OtorhinolaryngologyHead and Neck SurgeryHannover Medical SchoolHannoverGermany
| | - Jennifer Harre
- Department of OtorhinolaryngologyHead and Neck SurgeryHannover Medical SchoolHannoverGermany
| | - Hinrich Staecker
- Department of Otolaryngology, Head and Neck SurgeryUniversity of Kansas School of MedicineKansas CityKansas
| | - Nils Prenzler
- Department of OtorhinolaryngologyHead and Neck SurgeryHannover Medical SchoolHannoverGermany
| | - Dirk Strunk
- Institute of Experimental and Clinical Cell TherapySpinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical UniversitySalzburgAustria
| | - Sebastien Couillard‐Despres
- Institute of Experimental NeuroregenerationSpinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical UniversitySalzburgAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
| | - Pasquale Romanelli
- Institute of Experimental NeuroregenerationSpinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical UniversitySalzburgAustria
| | - Julia Hollerweger
- GMP Unit, Spinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
| | - Teresa Lassacher
- GMP Unit, Spinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
| | - Daniela Auer
- GMP Unit, Spinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
| | - Karin Pachler
- Research Program “Nanovesicular Therapies,”Paracelsus Medical University (PMU)SalzburgAustria
| | - Georg Wietzorrek
- Institute of Molecular and Cellular PharmacologyMedical University of InnsbruckInnsbruckAustria
| | - Ulrike Köhl
- Institute of Cellular TherapeuticsHannover Medical School and Clinical ImmunologyUniversity Leipzig, Fraunhofer Institute for Cell Therapy and ImmunologyLeipzigGermany
| | - Thomas Lenarz
- Department of OtorhinolaryngologyHead and Neck SurgeryHannover Medical SchoolHannoverGermany
| | - Katharina Schallmoser
- GMP Unit, Spinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
- Department of Transfusion MedicineUniversity HospitalSalzburger Landeskliniken GesmbH (SALK) and Paracelsus Medical University (PMU)SalzburgAustria
| | - Sandra Laner‐Plamberger
- Department of Transfusion MedicineUniversity HospitalSalzburger Landeskliniken GesmbH (SALK) and Paracelsus Medical University (PMU)SalzburgAustria
| | - Christine S. Falk
- Institute of Transplant ImmunologyHannover Medical SchoolHannoverGermany
| | - Eva Rohde
- GMP Unit, Spinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
- Department of Transfusion MedicineUniversity HospitalSalzburger Landeskliniken GesmbH (SALK) and Paracelsus Medical University (PMU)SalzburgAustria
| | - Mario Gimona
- GMP Unit, Spinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
- Research Program “Nanovesicular Therapies,”Paracelsus Medical University (PMU)SalzburgAustria
| |
Collapse
|
26
|
Chronic pain impact on rodents’ behavioral repertoire. Neurosci Biobehav Rev 2020; 119:101-127. [DOI: 10.1016/j.neubiorev.2020.09.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 07/14/2020] [Accepted: 09/21/2020] [Indexed: 12/20/2022]
|
27
|
Kimono D, Bose D, Seth RK, Mondal A, Saha P, Janulewicz P, Sullivan K, Lasley S, Horner R, Klimas N, Chatterjee S. Host Akkermansia muciniphila Abundance Correlates With Gulf War Illness Symptom Persistence via NLRP3-Mediated Neuroinflammation and Decreased Brain-Derived Neurotrophic Factor. Neurosci Insights 2020; 15:2633105520942480. [PMID: 32832901 PMCID: PMC7440889 DOI: 10.1177/2633105520942480] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 06/25/2020] [Indexed: 12/16/2022] Open
Abstract
Neurological disorders are commonly reported among veterans who returned from the Gulf war. Veterans who suffer from Gulf War illness (GWI) complain of continued symptom persistence that includes neurological disorders, muscle weakness, headaches, and memory loss, that developed during or shortly after the war. Our recent research showed that chemical exposure associated microbial dysbiosis accompanied by a leaky gut connected the pathologies in the intestine, liver, and brain. However, the mechanisms that caused the symptoms to persist even 30 years after the war remained elusive to investigators. In this study, we used a rodent model of GWI to investigate the persistence of microbiome alterations, resultant chronic inflammation, and its effect on neurotrophic and synaptic plasticity marker BDNF. The results showed that exposure to GW chemicals (the pesticide permethrin and prophylactic drug pyridostigmine bromide) resulted in persistent pathology characterized by the low relative abundance of the probiotic bacteria Akkermansia muciniphila in the gut, which correlated with high circulatory HMGB1 levels, blood-brain barrier dysfunction, neuroinflammation and lowered neurotrophin BDNF levels. Mechanistically, we used mice lacking the NLRP3 gene to investigate this inflammasome's role in observed pathology. These mice had significantly decreased inflammation and a subsequent increase in BDNF in the frontal cortex. This suggests that a persistently low species abundance of Akkermansia muciniphila and associated chronic inflammation due to inflammasome activation might be playing a significant role in contributing to chronic neurological problems in GWI. A therapeutic approach with various small molecules that can target both the restoration of a healthy microbiome and decreasing inflammasome activation might have better outcomes in treating GWI symptom persistence.
Collapse
Affiliation(s)
- Diana Kimono
- Environmental Health and Disease Laboratory, NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, USA
| | - Dipro Bose
- Environmental Health and Disease Laboratory, NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, USA
| | - Ratanesh K Seth
- Environmental Health and Disease Laboratory, NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, USA
| | - Ayan Mondal
- Environmental Health and Disease Laboratory, NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, USA
| | - Punnag Saha
- Environmental Health and Disease Laboratory, NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, USA
| | - Patricia Janulewicz
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Kimberly Sullivan
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Stephen Lasley
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA
| | - Ronnie Horner
- Department of Health Services Policy and Management, University of South Carolina, Columbia, SC, USA
| | - Nancy Klimas
- Department of Clinical Immunology, College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, NIEHS Center for Oceans and Human Health on Climate Change Interactions, Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
28
|
O'Reilly ML, Tom VJ. Neuroimmune System as a Driving Force for Plasticity Following CNS Injury. Front Cell Neurosci 2020; 14:187. [PMID: 32792908 PMCID: PMC7390932 DOI: 10.3389/fncel.2020.00187] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/29/2020] [Indexed: 12/15/2022] Open
Abstract
Following an injury to the central nervous system (CNS), spontaneous plasticity is observed throughout the neuraxis and affects multiple key circuits. Much of this spontaneous plasticity can elicit beneficial and deleterious functional outcomes, depending on the context of plasticity and circuit affected. Injury-induced activation of the neuroimmune system has been proposed to be a major factor in driving this plasticity, as neuroimmune and inflammatory factors have been shown to influence cellular, synaptic, structural, and anatomical plasticity. Here, we will review the mechanisms through which the neuroimmune system mediates plasticity after CNS injury. Understanding the role of specific neuroimmune factors in driving adaptive and maladaptive plasticity may offer valuable therapeutic insight into how to promote adaptive plasticity and/or diminish maladaptive plasticity, respectively.
Collapse
Affiliation(s)
- Micaela L O'Reilly
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Veronica J Tom
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
29
|
Sabaghi A, Heirani A, Kiani A, Yousofvand N, Sabaghi S. The Reduction of Seizure Intensity and Attenuation of Memory Deficiency and Anxiety-Like Behavior through Aerobic Exercise by Increasing the BDNF in Mice with Chronic Epilepsy. NEUROCHEM J+ 2020. [DOI: 10.1134/s1819712420020105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
30
|
Yang Y, Song Y, Zhang X, Zhao W, Ma T, Liu Y, Ma P, Zhao Y, Zhang H. Ketamine relieves depression-like behaviors induced by chronic postsurgical pain in rats through anti-inflammatory, anti-oxidant effects and regulating BDNF expression. Psychopharmacology (Berl) 2020; 237:1657-1669. [PMID: 32125485 DOI: 10.1007/s00213-020-05490-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 02/14/2020] [Indexed: 12/12/2022]
Abstract
RATIONALE Clinically, chronic postsurgical pain (CPSP) is very common. Many CPSP patients may experience depression. Thus far, little is known about the mechanism of the comorbidity of CPSP and depression. Ketamine has been confirmed to possess analgesic and rapid antidepressant effects, but it is unclear whether ketamine can relieve the comorbidity of CPSP and depression. OBJECTIVES The present study evaluated the effects of ketamine in rats with the comorbidity of CPSP and depression. METHODS We induced CPSP in rats by thoracotomy and screened for rats with or without depression-like phenotype by hierarchical cluster analysis based on the results of depression-related behavioral experiments. Subsequently, rats were intraperitoneally injected with ketamine (20 mg/kg) and were evaluated by mechanical withdrawal threshold, cold hyperalgesia test, sucrose preference test, forced swimming test, and open field test. The inflammatory-related cytokines (IL-1, IL-6, TNF-α, nuclear factor-kappaB), oxidative stress parameters (superoxide dismutase, malondialdehyde, glutathione, catalase), and brain-derived neurotrophic factor (BDNF) in rat hippocampus were detected. RESULTS In the hippocampus of rats with the comorbidity of CPSP and depression, IL-1, IL-6, TNF-α, nuclear factor-kappaB, and malondialdehyde were significantly increased, while superoxide dismutase, glutathione, catalase, and BDNF were significantly decreased. Ketamine relieved depression but did not attenuate hyperalgesia in CPSP rats. Additionally, ketamine reduced proinflammatory cytokines, inhibited oxidative stress, and elevated BDNF levels in rat hippocampus. CONCLUSIONS Ketamine can rapidly relieve CPSP-induced depression in rats, which may be related to the reduction of proinflammatory cytokines, regulating oxidative stress and increasing BDNF in the hippocampus.
Collapse
Affiliation(s)
- Yitian Yang
- Anesthesia and Operation Center, The First Medical Center of Chinese PLA General Hospital, Medical school of Chinese PLA, No. 28 Fuxing Road, Beijing, 100853, China.
| | - Yuxiang Song
- Anesthesia and Operation Center, The First Medical Center of Chinese PLA General Hospital, Medical school of Chinese PLA, No. 28 Fuxing Road, Beijing, 100853, China
| | - Xuan Zhang
- Department of Anesthesiology, Tianjin Cancer Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - Weixing Zhao
- Anesthesia and Operation Center, The First Medical Center of Chinese PLA General Hospital, Medical school of Chinese PLA, No. 28 Fuxing Road, Beijing, 100853, China
| | - Tao Ma
- Department of Anesthesiology, Rocket Army Characteristic Medical Center, Beijing, 100088, China
| | - Yi Liu
- Anesthesia and Operation Center, The First Medical Center of Chinese PLA General Hospital, Medical school of Chinese PLA, No. 28 Fuxing Road, Beijing, 100853, China
| | - Penglei Ma
- Department of Anesthesiology, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010030, China
| | - Yifan Zhao
- Department of Anesthesiology, The Fourth Medical Center of Chinese PLA General Hospital, Medical school of Chinese PLA, Beijing, 100037, China
| | - Hong Zhang
- Anesthesia and Operation Center, The First Medical Center of Chinese PLA General Hospital, Medical school of Chinese PLA, No. 28 Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
31
|
Guida F, De Gregorio D, Palazzo E, Ricciardi F, Boccella S, Belardo C, Iannotta M, Infantino R, Formato F, Marabese I, Luongo L, de Novellis V, Maione S. Behavioral, Biochemical and Electrophysiological Changes in Spared Nerve Injury Model of Neuropathic Pain. Int J Mol Sci 2020; 21:ijms21093396. [PMID: 32403385 PMCID: PMC7246983 DOI: 10.3390/ijms21093396] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/27/2020] [Accepted: 05/06/2020] [Indexed: 01/05/2023] Open
Abstract
Neuropathic pain is a pathological condition induced by a lesion or disease affecting the somatosensory system, with symptoms like allodynia and hyperalgesia. It has a multifaceted pathogenesis as it implicates several molecular signaling pathways involving peripheral and central nervous systems. Affective and cognitive dysfunctions have been reported as comorbidities of neuropathic pain states, supporting the notion that pain and mood disorders share some common pathogenetic mechanisms. The understanding of these pathophysiological mechanisms requires the development of animal models mimicking, as far as possible, clinical neuropathic pain symptoms. Among them, the Spared Nerve Injury (SNI) model has been largely characterized in terms of behavioral and functional alterations. This model is associated with changes in neuronal firing activity at spinal and supraspinal levels, and induces late neuropsychiatric disorders (such as anxious-like and depressive-like behaviors, and cognitive impairments) comparable to an advanced phase of neuropathy. The goal of this review is to summarize current findings in preclinical research, employing the SNI model as a tool for identifying pathophysiological mechanisms of neuropathic pain and testing pharmacological agent.
Collapse
Affiliation(s)
- Francesca Guida
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
- Correspondence: (F.G.); (S.M.)
| | - Danilo De Gregorio
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montréal, QC H3A1A1, Canada;
| | - Enza Palazzo
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Flavia Ricciardi
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Serena Boccella
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Carmela Belardo
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Monica Iannotta
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Rosmara Infantino
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Federica Formato
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Ida Marabese
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Livio Luongo
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Vito de Novellis
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Sabatino Maione
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
- Correspondence: (F.G.); (S.M.)
| |
Collapse
|
32
|
Zhang Z, Wu H, Liu Y, Gu X, Zhang W, Ma Z. The GCs-SGK1-ATP Signaling Pathway in Spinal Astrocytes Underlied Presurgical Anxiety-Induced Postsurgical Hyperalgesia. Anesth Analg 2020; 129:1163-1169. [PMID: 30113397 DOI: 10.1213/ane.0000000000003682] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Patients undergoing surgery often feel anxious. Accumulating evidence indicated that presurgical anxiety was related to the more severe postsurgical pain. An animal model was established that exposed Sprague-Dawley rats to a single-prolonged stress (SPS) procedure to induce presurgical anxiety-like behaviors. The experiment revealed that presurgical anxiety not only aggravated but also prolonged postsurgical pain. However, the underlying mechanisms were unknown. METHODS The rats in group C + Cort, group I + Cort, group A + Cort, and group AI + Cort were injected with corticosterone. The rats in group C + RU486, group I + RU486, group A + RU486, and group AI + RU486 were injected with mifepristone (RU486). The rats in group C + GSK650394 and group AI + GSK650394 were injected with GSK650394. The rats in group C + FC1 and group AI + FC1 were injected with fluorocitrate (FC) 30 minutes before SPS, 30 minutes before incision, and on postoperative days 1, 2, 3, 4, and 5. The rats in group C + FC2 and group AI + FC2 were injected with FC on postoperative days 7, 8, 9, 10, 11, 12, and 13. The paw withdrawal mechanical threshold was assessed 24 hours before SPS and from postoperative days 1 to 28. The level of corticosterone was determined by enzyme-linked immunosorbent assay. The expression of serum/glucocorticoid regulated kinase 1 (SGK1), interleukin-1β, and tumor necrosis factor-α was visualized by Western blot. The concentrations of adenosine triphosphate (ATP) were measured by ATP assay kit. RESULTS This study showed SPS elevated plasma glucocorticoids and ATP release from astrocytes, which meant the mechanical pain hypersensitivity in presurgical anxiety-induced postsurgical hyperalgesia was dependent on GCs-SGK1-ATP signaling pathway. SGK1 protein level in astrocytes was increased in response to the glucocorticoid stimuli and enhanced the extracellular release of ATP. Furthermore, spinal astrocytes played a key role in the maintenance. Targeting spinal astrocytes in maintenance phase prevented the pathological progression. CONCLUSIONS These data suggested an important signaling pathway that affected the pain sensitivity after operation caused by presurgical anxiety.
Collapse
Affiliation(s)
- ZuoXia Zhang
- From the Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu Province, China
| | | | | | | | | | | |
Collapse
|
33
|
Kremer M, Becker LJ, Barrot M, Yalcin I. How to study anxiety and depression in rodent models of chronic pain? Eur J Neurosci 2020; 53:236-270. [DOI: 10.1111/ejn.14686] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/06/2020] [Accepted: 01/14/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Mélanie Kremer
- Centre National de la Recherche Scientifique Institut des Neurosciences Cellulaires et Intégratives Université de Strasbourg Strasbourg France
| | - Léa J. Becker
- Centre National de la Recherche Scientifique Institut des Neurosciences Cellulaires et Intégratives Université de Strasbourg Strasbourg France
| | - Michel Barrot
- Centre National de la Recherche Scientifique Institut des Neurosciences Cellulaires et Intégratives Université de Strasbourg Strasbourg France
| | - Ipek Yalcin
- Centre National de la Recherche Scientifique Institut des Neurosciences Cellulaires et Intégratives Université de Strasbourg Strasbourg France
| |
Collapse
|
34
|
Brain-derived neurotrophic factor-TrkB signaling in the medial prefrontal cortex plays a role in the anhedonia-like phenotype after spared nerve injury. Eur Arch Psychiatry Clin Neurosci 2020; 270:195-205. [PMID: 29882089 PMCID: PMC7036057 DOI: 10.1007/s00406-018-0909-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 05/31/2018] [Indexed: 12/14/2022]
Abstract
Although depressive symptoms including anhedonia (i.e., loss of pleasure) frequently accompany pain, little is known about the risk factors contributing to individual differences in pain-induced anhedonia. In this study, we examined if signaling of brain-derived neurotrophic factor (BDNF) and its receptor tropomyosin-receptor-kinase B (TrkB) contribute to individual differences in the development of neuropathic pain-induced anhedonia. Rats were randomly subjected to spared nerved ligation (SNI) or sham surgery. The SNI rats were divided into two groups based on the results of a sucrose preference test. Rats with anhedonia-like phenotype displayed lower tissue levels of BDNF in the medial prefrontal cortex (mPFC) compared with rats without anhedonia-like phenotype and sham-operated rats. In contrast, tissue levels of BDNF in the nucleus accumbens (NAc) of rats with an anhedonia-like phenotype were higher compared with those of rats without anhedonia-like phenotype and sham-operated rats. Furthermore, tissue levels of BDNF in the hippocampus, L2-5 spinal cord, muscle, and liver from both rats with or without anhedonia-like phenotype were lower compared with those of sham-operated rats. A single injection of 7,8-dihydroxyflavone (10 mg/kg; TrkB agonist), but not ANA-12 (0.5 mg/kg; TrkB antagonist), ameliorated reduced sucrose preference and reduced BDNF-TrkB signaling in the mPFC in the rats with anhedonia-like phenotype. These findings suggest that reduced BDNF-TrkB signaling in the mPFC might contribute to neuropathic pain-induced anhedonia, and that TrkB agonists could be potential therapeutic drugs for pain-induced anhedonia.
Collapse
|
35
|
Polis AJ, Fitzgerald PJ, Hale PJ, Watson BO. Rodent ketamine depression-related research: Finding patterns in a literature of variability. Behav Brain Res 2019; 376:112153. [PMID: 31419519 PMCID: PMC6783386 DOI: 10.1016/j.bbr.2019.112153] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/11/2019] [Accepted: 08/13/2019] [Indexed: 12/23/2022]
Abstract
Discovering that the anesthetic drug ketamine has rapidly acting antidepressant effects in many individuals with major depression is one of the most important findings in clinical psychopharmacology in recent decades. The initial report of these effects in human subjects was based on a foundation of rodent preclinical studies carried out in the 1990s, and subsequent investigation has included both further studies in individuals with depression, as well as reverse translational experiments in animal models, especially rodents. While there is general agreement in the rodent literature that ketamine has rapidly-acting, and generally sustained, antidepressant-like properties, there are also points of contention across studies, including the precise mechanism of action of this drug. In this review, we briefly summarize prominent yet variable findings regarding the mechanism of action. We also discuss a combination of similarities and variances in the rodent literature in the antidepressant-like effects of ketamine as a function of dose, species and strain, test, stressor, and presumably sex of the experimenter. We then present previously unpublished mouse strain comparison data suggesting that subanesthetic ketamine does not have robust antidepressant-like properties in unstressed animals, and may actually promote depression-like behavior, in contrast to widely reported findings. We conclude that the data best support the notion of ketamine action principally via NMDA receptor antagonism, transiently boosting glutamatergic (and possibly other) signaling in diverse brain circuits. We also suggest that future studies should address in greater detail the extent to which antidepressant-like properties of this drug are stress-sensitive, in an effort to better model major depression present in humans.
Collapse
Affiliation(s)
- Andrew J Polis
- University of Michigan, Department of Psychiatry, Ann Arbor, MI 48109-5720, United States of America
| | - Paul J Fitzgerald
- University of Michigan, Department of Psychiatry, Ann Arbor, MI 48109-5720, United States of America
| | - Pho J Hale
- University of Michigan, Department of Psychiatry, Ann Arbor, MI 48109-5720, United States of America
| | - Brendon O Watson
- University of Michigan, Department of Psychiatry, Ann Arbor, MI 48109-5720, United States of America.
| |
Collapse
|
36
|
He L, Xu R, Chen Y, Liu X, Pan Y, Cao S, Xu T, Tian H, Zeng J. Intra-CA1 Administration of Minocycline Alters the Expression of Inflammation-Related Genes in Hippocampus of CCI Rats. Front Mol Neurosci 2019; 12:248. [PMID: 31708740 PMCID: PMC6822549 DOI: 10.3389/fnmol.2019.00248] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 09/26/2019] [Indexed: 11/13/2022] Open
Affiliation(s)
- Li He
- Department of Physiology, Zunyi Medical University, Zunyi, China
| | - Rui Xu
- Department of Physiology, Zunyi Medical University, Zunyi, China
| | - Yuanshou Chen
- Department of Physiology, Zunyi Medical University, Zunyi, China
| | - Xiaohong Liu
- Department of Physiology, Zunyi Medical University, Zunyi, China
| | - Youfu Pan
- Department of Genetics, Zunyi Medical University, Zunyi, China
| | - Song Cao
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical Univerisity, Zunyi, China
| | - Tao Xu
- Department of Physiology, Zunyi Medical University, Zunyi, China
| | - Hong Tian
- Department of Physiology, Zunyi Medical University, Zunyi, China
| | - Junwei Zeng
- Department of Physiology, Zunyi Medical University, Zunyi, China
| |
Collapse
|
37
|
Feng J, Wang M, Li M, Yang J, Jia J, Liu L, Zhou J, Zhang C, Wang X. Serum miR-221-3p as a new potential biomarker for depressed mood in perioperative patients. Brain Res 2019; 1720:146296. [PMID: 31211948 DOI: 10.1016/j.brainres.2019.06.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 06/10/2019] [Accepted: 06/14/2019] [Indexed: 01/04/2023]
Abstract
MicroRNAs (miRNAs) modulate various genes associated with brain disorders and circulating miRNAs may therefore serve as biomarkers for these neurological diseases. We previously found that the miRNA miR-221-3p was highly expressed in cerebrospinal fluid and the serum of major depressive disorder (MDD) patients. Here, we examined whether miR-221-3p could be used as a biomarker for depressed mood in perioperative patients. We first examined the relative expression of serum miR-221-3p by real-time quantitative PCR in perioperative patients with different degrees of depressive mood assessed by the Patient Health Questionnaire-9 (PHQ-9) diagnostic form. We found that miR-221-3p expression in the mild depressive mood group (PHQ-9 scores 5-9) was 2.21 fold that of the normal group (PHQ-9 scores 0-4) and the moderate&severe depressive mood group (PHQ-9 scores ≥ 10) showed miR-221-3p expression levels 3.66 fold that of the normal group. Then the absolute quantification of serum miR-221-3p was obtained using an miRNA standard curve. We found that the amount of serum miR-221-3p was positively correlated with depressed mood; when serum miR-221-3p > 1.7 × 107 copies/μg RNA, all indicated PHQ-9 scores were higher than 6. Subsequently, we found that miR-221-3p could indirectly increase the expression of IFN-α (Interferon alpha) in astrocytes by targeting IRF2 (Interferon Regulatory Factor 2) and that miR-221-3p participated in the anti-neuroinflammatory signaling cascades induced by ketamine and paroxetine via the IRF2/IFN-α pathway. Our results indicate that elevated serum miR-221-3p can be used as a biomarker for depressed mood in perioperative patients and that IFN-α-induced NF-κB activation in astrocytes mediated by miR-221-3p targeting of IRF2 may be one of the potential mechanisms.
Collapse
Affiliation(s)
- Jianguo Feng
- Laboratory of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Maozhou Wang
- Department of Intensive Care Unit, The Affiliated Chaoyang Hospital of Capital Medical University, Beijing, China
| | - Mao Li
- Laboratory of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jimei Yang
- Laboratory of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jing Jia
- Laboratory of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Li Liu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jun Zhou
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Chunxiang Zhang
- Department of Biomedical Engineering, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Xiaobin Wang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China.
| |
Collapse
|
38
|
Robertson OD, Coronado NG, Sethi R, Berk M, Dodd S. Putative neuroprotective pharmacotherapies to target the staged progression of mental illness. Early Interv Psychiatry 2019; 13:1032-1049. [PMID: 30690898 DOI: 10.1111/eip.12775] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/26/2018] [Indexed: 12/22/2022]
Abstract
AIM Neuropsychiatric disorders including depression, bipolar and schizophrenia frequently exhibit a neuroprogressive course from prodrome to chronicity. There are a range of agents exhibiting capacity to attenuate biological mechanisms associated with neuroprogression. This review will update the evidence for putative neuroprotective agents including clinical efficacy, mechanisms of action and limitations in current assessment tools, and identify novel agents with neuroprotective potential. METHOD Data for this review were sourced from online databases PUBMED, Embase and Web of Science. Only data published since 2012 were included in this review, no data were excluded based on language or publication origin. RESULTS Each of the agents reviewed inhibit one or multiple pathways of neuroprogression including: inflammatory gene expression and cytokine release, oxidative and nitrosative stress, mitochondrial dysfunction, neurotrophin dysregulation and apoptotic signalling. Some demonstrate clinical efficacy in preventing neural damage or loss, relapse or cognitive/functional decline. Agents include: the psychotropic medications lithium, second generation antipsychotics and antidepressants; other pharmacological agents such as minocycline, aspirin, cyclooxygenase-2 inhibitors, statins, ketamine and alpha-2-delta ligands; and others such as erythropoietin, oestrogen, leptin, N-acetylcysteine, curcumin, melatonin and ebselen. CONCLUSIONS Signals of evidence of clinical neuroprotection are evident for a number of candidate agents. Adjunctive use of multiple agents may present a viable avenue to clinical realization of neuroprotection. Definitive prospective studies of neuroprotection with multimodal assessment tools are required.
Collapse
Affiliation(s)
- Oliver D Robertson
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia.,Mental Health, Drugs and Alcohol Services, University Hospital Geelong, Barwon Health, Geelong, Victoria, Australia
| | - Nieves G Coronado
- Unidad de Gestión Clinica Salud Mental, Hospital Universitario Virgen del Rocio, Sevilla, Spain
| | - Rickinder Sethi
- Department of Psychiatry, Western University, London, Ontario, Canada
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia.,Mental Health, Drugs and Alcohol Services, University Hospital Geelong, Barwon Health, Geelong, Victoria, Australia.,Department of Psychiatry, The University of Melbourne, Parkville, Victoria, Australia.,Mood Disorders Research Program, Orygen, the National Centre of Excellence in Youth Mental Health, Parkville, Victoria, Australia.,Department of Psychiatry, Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Seetal Dodd
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia.,Mental Health, Drugs and Alcohol Services, University Hospital Geelong, Barwon Health, Geelong, Victoria, Australia.,Department of Psychiatry, The University of Melbourne, Parkville, Victoria, Australia.,Mood Disorders Research Program, Orygen, the National Centre of Excellence in Youth Mental Health, Parkville, Victoria, Australia
| |
Collapse
|
39
|
Temporal changes in physiological and molecular markers in various brain regions following transient global ischemia in rats. Mol Biol Rep 2019; 46:6215-6230. [PMID: 31576510 DOI: 10.1007/s11033-019-05060-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 08/31/2019] [Indexed: 12/11/2022]
Abstract
Several mechanisms are involved in the loss of cellular integrity and tissue destructions in various brain regions during ischemic insult. The affected brain employs various self-repair mechanisms during the poststroke recovery. Therefore, the current study involves time course changes in different brain regions following ischemia in terms of inflammation, oxidative stress and apoptosis for which a bilateral common carotid arteries occlusion model was chosen. The development of oxidative stress was seen with a marked increase in ROS and NO levels with concomitant decrease in GSH levels and also the activities of anti-oxidant enzymes. These alterations were accompanied with decreased levels of neurotransmitters and motor and cognitive deficits at various time points. Increased expressions of various pro-inflammatory cytokines and a decline in BDNF levels in hippocampal regions on 7th day post ischemia, suggesting their role in its pathogenesis. The restoration of BDNF and neurotransmitter levels along with significant decline in inflammatory cytokine levels 14th day onwards following ischemia in hippocampus suggested poststroke recovery. The extent of neuronal damage was found to be increased significantly on 7th day post ischemia as indicated by TUNEL assay and hematoxylin and eosin staining depicting enhanced number of pyknotic neurons in cortical and hippocampal regions. Cortical regions of the ischemic brains were severely affected while hippocampal regions showed significant poststroke recovery, which might attributed to the normalization of BDNF and pro-inflammatory cytokine levels. In conclusion, the present study established the central role of BDNF and pro-inflammatory cytokines in the poststroke recovery. Also, the cortical and hippocampal regions were found to be more susceptible for ischemic injury. As our results indicated, full recovery after ischemic injury in different brain regions was not achieved, therefore further studies with long-term recovery time are required to be conducted.
Collapse
|
40
|
Fiore NT, Austin PJ. Peripheral Nerve Injury Triggers Neuroinflammation in the Medial Prefrontal Cortex and Ventral Hippocampus in a Subgroup of Rats with Coincident Affective Behavioural Changes. Neuroscience 2019; 416:147-167. [DOI: 10.1016/j.neuroscience.2019.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 06/25/2019] [Accepted: 08/02/2019] [Indexed: 12/14/2022]
|
41
|
Smalheiser NR. Ketamine: A Neglected Therapy for Alzheimer Disease. Front Aging Neurosci 2019; 11:186. [PMID: 31396078 PMCID: PMC6667975 DOI: 10.3389/fnagi.2019.00186] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 07/09/2019] [Indexed: 01/05/2023] Open
Affiliation(s)
- Neil R Smalheiser
- Department of Psychiatry, Psychiatric Institute, University of Illinois School of Medicine, Chicago, IL, United States
| |
Collapse
|
42
|
Fang X, Zhan G, Zhang J, Xu H, Zhu B, Hu Y, Yang C, Luo A. Abnormalities in Inflammatory Cytokines Confer Susceptible to Chronic Neuropathic Pain-related Anhedonia in a Rat Model of Spared Nerve Injury. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2019; 17:189-199. [PMID: 30905119 PMCID: PMC6478091 DOI: 10.9758/cpn.2019.17.2.189] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/31/2018] [Accepted: 02/05/2018] [Indexed: 12/15/2022]
Abstract
Objective Patients with chronic neuropathic pain (CNP) have a higher incidence to develop depression. However, its pathogenesis has not yet been fully elucidated. Here we aimed to investigate the role of inflammatory cytokines in CNP-related anhedonia, which is a core symptom of depression, and to explore the effects of ketamine and parecoxib on pain and anhedonia. Methods A rat model of spared nerve injury (SNI) was constructed to mimic CNP. Hierarchical cluster analysis of sucrose preference test (SPT) was applied to classify the SNI rats into anhedonia susceptible and unsusceptible. Inflammatory cytokines in medial prefrontal cortex (mPFC) of brain, serum and L2–5 spinal cord were measured. Moreover, effects of ketamine or parecoxib on mechanical withdrawal test (MWT) and SPT in anhedonia susceptible rats were detected. Results Tumor necrosis factor (TNF)-α was increased in mPFC, serum and and spinal cord of anhedonia susceptible rats. Furthermore, anhedonia susceptible and unsusceptible rats both increased the interleukin (IL)-1β level in mPFC, serum and spinal cord. IL-6 was altered in serum and spinal cord, but not in mPFC. IL-10 was significantly altered in mPFC and serum, but not in spinal cord. Additionally, ketamine treatment significantly attenuated the decreased results of MWT and SPT in anhedonia susceptible rats, and that parecoxib significantly improved the MWT score, but failed to alter the result of SPT. Conclusion These findings suggest that abnormalities in inflammatory cytokines confer susceptible to anhedonia in a rat model of SNI. Ketamine, a fast-acting antidepressant, has pharmacological benefits to alleviate pain and anhedonia symptoms.
Collapse
Affiliation(s)
- Xi Fang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gaofeng Zhan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Xu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Zhu
- Department of Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yimin Hu
- Department of Anestesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chun Yang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ailin Luo
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
43
|
Corriger A, Duclos M, Corcuff JB, Lambert C, Marceau G, Sapin V, Macian N, Roux D, Pereira B, Pickering G. Hormonal Status and Cognitivo-Emotional Profile in Real-Life Patients With Neuropathic Pain: A Case Control Study. Pain Pract 2019; 19:703-714. [PMID: 31127700 DOI: 10.1111/papr.12800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 05/17/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND The specific impact of neuropathic pain and recommended neuropathic pain treatments on the hormonal and immune status of patients has been so far poorly explored. This study aimed at studying, in real life, the hypothalamic-pituitary-adrenal axis and the cytokine profile of patients with neuropathic pain. It also explored their links with cognition, emotion, quality of life, and drug treatment. METHODS This prospective study (clinicaltrials.gov NCT01543425) included 60 patients with neuropathic pain and 60 age- and gender-matched healthy volunteers after obtaining signatures of informed consent. A number of parameters were measured: adrenocorticotropic hormone, cortisol, cortisol awakening response, dehydroepiandrosterone sulphate, sex hormone binding globulin, testosterone, 17-β-estradiol, progesterone, luteinizing hormone, follicle-stimulating hormone, cytokines, brain-derived neurotrophic factor, and vitamin D. Psychological parameters were assessed by questionnaires. RESULTS Patients with neuropathic pain had lower levels of adrenocorticotropic hormone (P = 0.009) and dehydroepiandrosterone sulphate (P < 0.001) than controls, and the cortisol awakening response was impaired. Patients were more depressed and anxious (P < 0.001) and had a diminished quality of life (P < 0.001), which was influenced by cytokines (P = 0.0067) and testosterone (P = 0.028). Antidepressants and antiepileptics appeared to interfere with testosterone and cognitivo-emotional domains. CONCLUSION An impairment of the hormonal status and of the immune system was observed in patients. It identified testosterone as a potential pivotal mediator between antidepressants/antiepileptics and quality of life. Further studies must address the exact impact of different types of drugs on central effects, of gender differences, and of the immune system of neuropathic pain.
Collapse
Affiliation(s)
- Alexandrine Corriger
- Laboratoire Neuro-Dol Inserm 1107, University Clermont Auvergne, Clermont-Ferrand, France
| | - Martine Duclos
- Sports Medicine Department, University Hospital, CHU, INRA UMR 1019, UNH, CRNH, Clermont-Ferrand, France
| | - Jean-Benoit Corcuff
- Hormone Laboratory, Nuclear Medicine, CHU Bordeaux UMR INRA 1286 - University Bordeaux, Bordeaux, France
| | - Céline Lambert
- Biostatistics Unit (DRCI), University Hospital Clermont-Ferrand, Clermont-Ferrand, France
| | - Geoffroy Marceau
- Biochemistry Department, University Hospital CHU, Clermont-Ferrand, France
| | - Vincent Sapin
- Biochemistry Department, University Hospital CHU, Clermont-Ferrand, France
| | - Nicolas Macian
- Clinical Pharmacology Department, CPC/CIC Inserm 1405, University Hospital CHU, Clermont-Ferrand, France
| | - Delphine Roux
- Clinical Pharmacology Department, CPC/CIC Inserm 1405, University Hospital CHU, Clermont-Ferrand, France
| | - Bruno Pereira
- Biostatistics Unit (DRCI), University Hospital Clermont-Ferrand, Clermont-Ferrand, France
| | - Gisèle Pickering
- Laboratoire Neuro-Dol Inserm 1107, University Clermont Auvergne, Clermont-Ferrand, France.,Clinical Pharmacology Department, CPC/CIC Inserm 1405, University Hospital CHU, Clermont-Ferrand, France
| |
Collapse
|
44
|
Jang HM, Lee KE, Kim DH. The Preventive and Curative Effects of Lactobacillus reuteri NK33 and Bifidobacterium adolescentis NK98 on Immobilization Stress-Induced Anxiety/Depression and Colitis in Mice. Nutrients 2019; 11:nu11040819. [PMID: 30979031 PMCID: PMC6521032 DOI: 10.3390/nu11040819] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/03/2019] [Accepted: 04/09/2019] [Indexed: 01/01/2023] Open
Abstract
The gut dysbiosis by stressors such as immobilization deteriorates psychiatric disorders through microbiota-gut-brain axis activation. To understand whether probiotics could simultaneously alleviate anxiety/depression and colitis, we examined their effects on immobilization stress (IS)-induced anxiety/depression and colitis in mice. The probiotics Lactobacillus reuteri NK33 and Bifidobacterium adolescentis NK98 were isolated from healthy human feces. Mice with anxiety/depression and colitis were prepared by IS treatment. NK33 and NK98 potently suppressed NF-κB activation in lipopolysaccharide (LPS)-induced BV-2 cells. Treatment with NK33 and/or NK98, which were orally gavaged in mice before or after IS treatment, significantly suppressed the occurrence and development of anxiety/depression, infiltration of Iba1+ and LPS+/CD11b+ cells (activated microglia) into the hippocampus, and corticosterone, IL-6, and LPS levels in the blood. Furthermore, they induced hippocampal BDNF expression while NF-κB activation was suppressed. NK33 and/or NK98 treatments suppressed IS-induced colon shortening, myeloperoxidase activity, infiltration of CD11b+/CD11c+ cells, and IL-6 expression in the colon. Their treatments also suppressed the IS-induced fecal Proteobacteria population and excessive LPS production. They also induced BDNF expression in LPS-induced SH-SY5Y cells in vitro. In conclusion, NK33 and NK98 synergistically alleviated the occurrence and development of anxiety/depression and colitis through the regulation of gut immune responses and microbiota composition.
Collapse
Affiliation(s)
- Hyo-Min Jang
- Neurobiota Research Center, Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Kyung-Eon Lee
- Neurobiota Research Center, Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Dong-Hyun Kim
- Neurobiota Research Center, Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| |
Collapse
|
45
|
Humo M, Lu H, Yalcin I. The molecular neurobiology of chronic pain-induced depression. Cell Tissue Res 2019; 377:21-43. [PMID: 30778732 DOI: 10.1007/s00441-019-03003-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 02/01/2019] [Indexed: 12/18/2022]
Abstract
The increasing number of individuals with comorbidities poses an urgent need to improve the management of patients with multiple co-existing diseases. Among these comorbidities, chronic pain and mood disorders, two long-lasting disabling conditions that significantly reduce the quality of life, could be cited first. The recent development of animal models accelerated the studies focusing on the underlying mechanisms of the chronic pain and depression/anxiety comorbidity. This review provides an overview of clinical and pre-clinical studies performed over the past two decades addressing the molecular aspects of the comorbid relationship of chronic pain and depression. We thus focused on the studies that investigated the molecular characteristics of the comorbid relationship between chronic pain and mood disorders, especially major depressive disorders, from the genetic and epigenetic point of view to key neuromodulators which have been shown to play an important role in this comorbidity.
Collapse
Affiliation(s)
- Muris Humo
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique et Université de Strasbourg, 67000, Strasbourg, France
| | - Han Lu
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique et Université de Strasbourg, 67000, Strasbourg, France.,Faculty of Biology and Bernstein Center Freiburg, University of Freiburg, D-79104, Freiburg, Germany
| | - Ipek Yalcin
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique et Université de Strasbourg, 67000, Strasbourg, France.
| |
Collapse
|
46
|
Yang C, Fang X, Zhan G, Huang N, Li S, Bi J, Jiang R, Yang L, Miao L, Zhu B, Luo A, Hashimoto K. Key role of gut microbiota in anhedonia-like phenotype in rodents with neuropathic pain. Transl Psychiatry 2019; 9:57. [PMID: 30705252 PMCID: PMC6355832 DOI: 10.1038/s41398-019-0379-8] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 12/15/2018] [Accepted: 01/02/2019] [Indexed: 02/07/2023] Open
Abstract
Patients with chronic neuropathic pain frequently suffer from symptoms of anhedonia, which is a core symptom of depression. Accumulating studies suggest that gut microbiota may play a role in depression via gut-microbiota-brain axis. However, it is unknown whether gut microbiota plays a role in neuropathic pain-associated anhedonia. Here, we used a rat model of spared nerve injury (SNI). Hierarchical cluster analysis of sucrose preference test (SPT) results was used to classify the SNI rats with or without anhedonia-like phenotype. The 16S ribosomal RNA sequencing analysis showed abnormal composition of gut microbiota in the anhedonia susceptible compared to sham-operated rats and resilient rats. Furthermore, antibiotics-treated mice showed pain as well as depression-like and anhedonia-like phenotypes, suggesting a role of gut microbiota in these abnormal behaviors. Transplantation of fecal microbiota from anhedonia susceptible rats into antibiotics-treated pseudo-germ-free mice significantly exaggerated pain and depression-like phenotypes, including anhedonia. In contrast, transplantation of fecal microbiota from resilient rats into antibiotics-treated pseudo-germ-free mice significantly improved pain and depression-like phenotypes, including anhedonia. In conclusion, this study suggests that abnormal composition of gut microbiota may contribute to anhedonia susceptibility post SNI surgery, and that gut microbiota also plays a role in the pain as well as depression-like phenotypes. Interestingly, fecal microbiota transplantation from SNI rats with or without anhedonia can alter pain, depression-like and anhedonia-like phenotypes in the pseudo-germ-free mice. Therefore, it is likely that gut microbiota plays a key role in the pain as well as depression-like phenotypes including anhedonia in rodents with neuropathic pain.
Collapse
Affiliation(s)
- Chun Yang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Fang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gaofeng Zhan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Niannian Huang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiangjiang Bi
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Riyue Jiang
- Department of Internal Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Ling Yang
- Department of Internal Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Liying Miao
- Department of Internal Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Bin Zhu
- Department of Internal Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Ailin Luo
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan.
| |
Collapse
|
47
|
Fiore NT, Austin PJ. Glial-cytokine-neuronal Adaptations in the Ventral Hippocampus of Rats with Affective Behavioral Changes Following Peripheral Nerve Injury. Neuroscience 2018; 390:119-140. [DOI: 10.1016/j.neuroscience.2018.08.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/01/2018] [Accepted: 08/07/2018] [Indexed: 12/17/2022]
|
48
|
Kummer KK, Kalpachidou T, Mitrić M, Langeslag M, Kress M. Altered Gene Expression in Prefrontal Cortex of a Fabry Disease Mouse Model. Front Mol Neurosci 2018; 11:201. [PMID: 30013462 PMCID: PMC6036252 DOI: 10.3389/fnmol.2018.00201] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/18/2018] [Indexed: 11/13/2022] Open
Abstract
Fabry disease is an X-chromosome linked hereditary disease that is caused by loss of function mutations in the α-galactosidase A (α-Gal A) gene, resulting in defective glycolipid degradation and subsequent accumulation of globotriaosylceramide (Gb3) in different tissues, including vascular endothelial cells and neurons in the peripheral and central nervous system. We recently reported a differential gene expression profile of α-Gal A(−/0) mouse dorsal root ganglia, an established animal model of Fabry disease, thereby providing new gene targets that might underlie the neuropathic pain related symptoms. To investigate the cognitive symptoms experienced by Fabry patients, we performed one-color based hybridization microarray expression profiling of prefrontal cortex samples from adult α-Gal A(−/0) mice and age-matched wildtype controls, followed by protein-protein interaction and pathway analyses for the differentially regulated mRNAs. We found that from a total of 381 differentially expressed genes, 135 genes were significantly upregulated, whereas 246 genes were significantly downregulated between α-Gal A(−/0) mice and wildtype controls. Enrichment analysis for downregulated genes revealed mainly immune related pathways, including immune/defense responses, regulation of cytokine production, as well as signaling and transport regulation pathways. Further analysis of the regulated genes revealed a large number of genes involved in neurodegeneration. The current analysis for the first time presents a differential gene expression profile of central nervous system tissue from α-Gal A(−/0) mice, thereby providing novel knowledge on the deregulation and a possible contribution of gene expression to Fabry disease related brain pathologies.
Collapse
Affiliation(s)
- Kai K Kummer
- Division of Physiology, Department of Physiology and Medical Physics Medical, University of Innsbruck, Innsbruck, Austria
| | - Theodora Kalpachidou
- Division of Physiology, Department of Physiology and Medical Physics Medical, University of Innsbruck, Innsbruck, Austria
| | - Miodrag Mitrić
- Division of Physiology, Department of Physiology and Medical Physics Medical, University of Innsbruck, Innsbruck, Austria
| | - Michiel Langeslag
- Division of Physiology, Department of Physiology and Medical Physics Medical, University of Innsbruck, Innsbruck, Austria
| | - Michaela Kress
- Division of Physiology, Department of Physiology and Medical Physics Medical, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
49
|
Ong WY, Stohler CS, Herr DR. Role of the Prefrontal Cortex in Pain Processing. Mol Neurobiol 2018; 56:1137-1166. [PMID: 29876878 PMCID: PMC6400876 DOI: 10.1007/s12035-018-1130-9] [Citation(s) in RCA: 413] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 05/14/2018] [Indexed: 12/16/2022]
Abstract
The prefrontal cortex (PFC) is not only important in executive functions, but also pain processing. The latter is dependent on its connections to other areas of the cerebral neocortex, hippocampus, periaqueductal gray (PAG), thalamus, amygdala, and basal nuclei. Changes in neurotransmitters, gene expression, glial cells, and neuroinflammation occur in the PFC during acute and chronic pain, that result in alterations to its structure, activity, and connectivity. The medial PFC (mPFC) could serve dual, opposing roles in pain: (1) it mediates antinociceptive effects, due to its connections with other cortical areas, and as the main source of cortical afferents to the PAG for modulation of pain. This is a ‘loop’ where, on one side, a sensory stimulus is transformed into a perceptual signal through high brain processing activity, and perceptual activity is then utilized to control the flow of afferent sensory stimuli at their entrance (dorsal horn) to the CNS. (2) It could induce pain chronification via its corticostriatal projection, possibly depending on the level of dopamine receptor activation (or lack of) in the ventral tegmental area-nucleus accumbens reward pathway. The PFC is involved in biopsychosocial pain management. This includes repetitive transcranial magnetic stimulation, transcranial direct current stimulation, antidepressants, acupuncture, cognitive behavioral therapy, mindfulness, music, exercise, partner support, empathy, meditation, and prayer. Studies demonstrate the role of the PFC during placebo analgesia, and in establishing links between pain and depression, anxiety, and loss of cognition. In particular, losses in PFC grey matter are often reversible after successful treatment of chronic pain.
Collapse
Affiliation(s)
- Wei-Yi Ong
- Department of Anatomy, National University of Singapore, Singapore, 119260, Singapore.
- Neurobiology and Ageing Research Programme, National University of Singapore, Singapore, 119260, Singapore.
| | | | - Deron R Herr
- Department of Pharmacology, National University of Singapore, Singapore, 119260, Singapore.
| |
Collapse
|
50
|
Cho C, Michailidis V, Martin LJ. Revealing brain mechanisms of mTOR-mediated translational regulation: Implications for chronic pain. NEUROBIOLOGY OF PAIN 2018; 4:27-34. [PMID: 31194026 PMCID: PMC6550104 DOI: 10.1016/j.ynpai.2018.03.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/15/2018] [Accepted: 03/16/2018] [Indexed: 12/27/2022]
Abstract
mTOR is a major regulator of protein translation. mTOR serves an important role in neural plasticity. mTOR signalling in the brain as a pathology for neurological disorder is known. mTOR signalling in the brain as a chronic pain mechanism is understudied.
In the spinal cord, altered protein transcription and translation have received a lot of recent attention for their role in neural plasticity, a major mechanism leading to the development of chronic pain. However, changes in brain plasticity are also associated with the maintenance of pain symptoms, but these cellular mechanisms remain less clear. The mechanistic/mammalian target of rapamycin (mTOR) is a master regulator of protein synthesis, and controls several neuronal functions, including neural plasticity. While aberrant changes in mTOR signaling are associated with sensitization of the pain pathway (sensory neurons and spinal cord), there are various nervous system diseases that have pain as a comorbidity and altered mTOR activity in the brain. Here, we provide a brief review of mTOR changes in the brain that are associated with some neurological disorders and focus on how these changes may be relevant to the pain of the underlying condition and chronic pain itself.
Collapse
Affiliation(s)
- Chulmin Cho
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
| | - Vassilia Michailidis
- Deptartment of Cell and Systems Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
| | - Loren J. Martin
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
- Deptartment of Cell and Systems Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
- Corresponding author at: Department of Psychology, University of Toronto Mississauga, 3359 Mississauga Rd., Mississauga, ON L5L 1C6, Canada.
| |
Collapse
|