1
|
Pei Y, Liu H, Lang J, Chen Y, Zhang F, Hao R, Li J, Gu S, Peng Q, Song J, Zhang Z. rTMS ameliorates CUMS-induced anxiety-depression-like behaviour and cognitive dysfunction in rats by modulating the COX-2/PGE2 signalling pathway. J Psychiatr Res 2025; 186:116-128. [PMID: 40233438 DOI: 10.1016/j.jpsychires.2025.04.008] [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: 01/17/2025] [Revised: 03/30/2025] [Accepted: 04/08/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND rTMS is a safe and effective neuromodulation method for treating depression, but the specifics of its antidepressant effects and the underlying mechanisms remain uncertain. METHODS Male SD rats were randomly divided into four groups: control group, CUMS group, CUMS + rTMS (10 Hz) group, and CUMS + celecoxib (25 mg/kg, as a positive control) group. Depression-like behavior was assessed by weight change, SPT, and FST; anxiety by OFT and EPM; and cognitive function by the Y-maze. WB, IF, ELISA, and qPCR were used to observe changes in COX-2/PGE2 signaling pathway-related proteins, inflammatory factors, and the activation of astrocytes and microglia in the hippocampus of rats. RESULTS Compared to the control group, rats in the CUMS group exhibited significant anxiety-depression-like behavior and cognitive dysfunction. Compared to the CUMS group, rTMS and celecoxib interventions improved anxiety-depression-like behavior and cognitive dysfunction, reduced the expression of microglia and astrocytes, reversed the upregulation of pro-inflammatory factors (IL-1β, IL-6, TNF-α), and downregulated the expression of proteins related to the COX-2/PGE2 signaling pathway in CUMS-induced rats. CONCLUSIONS The study demonstrated that rTMS could improve anxiety-depression-like behavior and cognitive dysfunction in rats by modulating the COX-2/PGE2 pathway.
Collapse
Affiliation(s)
- Yanjiao Pei
- Henan Key Laboratory of Biological Psychiatry, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453002, China
| | - Huanhuan Liu
- Henan Key Laboratory of Biological Psychiatry, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453002, China; Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453002, China
| | - Jiqing Lang
- Henan Key Laboratory of Biological Psychiatry, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453002, China
| | - Yuxin Chen
- Henan Key Laboratory of Biological Psychiatry, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453002, China
| | - Fuping Zhang
- Henan Key Laboratory of Biological Psychiatry, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453002, China; Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453002, China; Brain Institute, Henan Academy of Innovations in Medical Science, China; Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, the Second Affiliated Hospital of Xinxiang Medical University, China
| | - Ran Hao
- Jinan Mental Health Center, Jinan, Shandong, 250309, China
| | - Jiao Li
- The First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Neurorestoratology, Weihai, Henan, 453100, China
| | - Shina Gu
- The First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Neurorestoratology, Weihai, Henan, 453100, China
| | - Qi Peng
- Xinxiang Medical University, Xinxiang, Henan, 453000, China
| | - Jinggui Song
- Henan Key Laboratory of Biological Psychiatry, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453002, China; Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453002, China.
| | - Zhaohui Zhang
- The First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Neurorestoratology, Weihai, Henan, 453100, China.
| |
Collapse
|
2
|
Katsipis G, Lavrentiadou SN, Geromichalos GD, Tsantarliotou MP, Halevas E, Litsardakis G, Pantazaki AA. Evaluation of the Anti-Amyloid and Anti-Inflammatory Properties of a Novel Vanadium(IV)-Curcumin Complex in Lipopolysaccharides-Stimulated Primary Rat Neuron-Microglia Mixed Cultures. Int J Mol Sci 2024; 26:282. [PMID: 39796150 PMCID: PMC11720140 DOI: 10.3390/ijms26010282] [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: 11/18/2024] [Revised: 12/22/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Lipopolysaccharides (LPS) are bacterial mediators of neuroinflammation that have been detected in close association with pathological protein aggregations of Alzheimer's disease. LPS induce the release of cytokines by microglia and mediate the upregulation of inducible nitric oxide synthase (iNOS)-a mechanism also associated with amyloidosis. Curcumin is a recognized natural medicine but has extremely low bioavailability. V-Cur, a novel hemocompatible Vanadium(IV)-curcumin complex with higher solubility and bioactivity than curcumin, is studied here. Co-cultures consisting of rat primary neurons and microglia were treated with LPS and/or curcumin or V-Cur. V-Cur disrupted LPS-induced overexpression of amyloid precursor protein (APP) and the in vitro aggregation of human insulin (HI), more effectively than curcumin. Cell stimulation with LPS also increased full-length, inactive, and total iNOS levels, and the inflammation markers IL-1β and TNF-α. Both curcumin and V-Cur alleviated these effects, with V-Cur reducing iNOS levels more than curcumin. Complementary insights into possible bioactivity mechanisms of both curcumin and V-Cur were provided by In silico molecular docking calculations on Aβ1-42, APP, Aβ fibrils, HI, and iNOS. This study renders curcumin-based compounds a promising anti-inflammatory intervention that may be proven a strong tool in the effort to mitigate neurodegenerative disease pathology and neuroinflammatory conditions.
Collapse
Affiliation(s)
- Georgios Katsipis
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (G.K.); (E.H.)
- Center for Interdisciplinary Research and Innovation, Laboratory of Neurodegenerative Diseases (LND), Thermi, 57001 Thessaloniki, Greece;
| | - Sophia N. Lavrentiadou
- Center for Interdisciplinary Research and Innovation, Laboratory of Neurodegenerative Diseases (LND), Thermi, 57001 Thessaloniki, Greece;
- Laboratory of Animal Physiology, School of Veterinary Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - George D. Geromichalos
- Center for Interdisciplinary Research and Innovation, Laboratory of Neurodegenerative Diseases (LND), Thermi, 57001 Thessaloniki, Greece;
- Laboratory of Inorganic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Maria P. Tsantarliotou
- Laboratory of Animal Physiology, School of Veterinary Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Eleftherios Halevas
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (G.K.); (E.H.)
- Institute of Biosciences & Applications, National Centre for Scientific Research “Demokritos”, 15310 Athens, Greece
| | - George Litsardakis
- Laboratory of Materials for Electrotechnics, School of Electrical and Computer Engineering, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Anastasia A. Pantazaki
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (G.K.); (E.H.)
- Center for Interdisciplinary Research and Innovation, Laboratory of Neurodegenerative Diseases (LND), Thermi, 57001 Thessaloniki, Greece;
| |
Collapse
|
3
|
Hall S, Parr BA, Hussey S, Anoopkumar-Dukie S, Arora D, Grant GD. The neurodegenerative hypothesis of depression and the influence of antidepressant medications. Eur J Pharmacol 2024; 983:176967. [PMID: 39222740 DOI: 10.1016/j.ejphar.2024.176967] [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: 07/04/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Depression is a complex neurological disease that holds many theories on its aetiology and pathophysiology. The monoamine strategy of treating depression with medications to increase levels of monoamines in the (extra)synapse, primarily through the inhibition of monoamine transporters, does not always work, as seen in patients that lack a response to multiple anti-depressant exposures, as well as a lack of depressive symptoms in healthy volunteers exposed to monoamine reduction. Depression is increasingly being understood not as a single condition, but as a complex interplay of adaptations in various systems, including inflammatory responses and neurotransmission pathways in the brain. This understanding has led to the development of the neurodegenerative hypothesis of depression. This hypothesis, which is gaining widespread acceptance posits that both oxidative stress and inflammation play significant roles in the pathophysiology of depression. This article is a review of the literature focused on neuroinflammation in depression, as well as summarised studies of anti-inflammatory and antioxidant effects of antidepressants.
Collapse
Affiliation(s)
- Susan Hall
- School of Pharmacy and Medical Sciences, Griffith University Gold Coast Campus, Southport, 4222, Australia.
| | - Brie-Anne Parr
- School of Pharmacy and Medical Sciences, Griffith University Gold Coast Campus, Southport, 4222, Australia
| | - Sarah Hussey
- School of Pharmacy and Medical Sciences, Griffith University Gold Coast Campus, Southport, 4222, Australia
| | | | - Devinder Arora
- School of Pharmacy and Medical Sciences, Griffith University Gold Coast Campus, Southport, 4222, Australia
| | - Gary D Grant
- School of Pharmacy and Medical Sciences, Griffith University Gold Coast Campus, Southport, 4222, Australia
| |
Collapse
|
4
|
De Lott LB, Khan N, Woodward MA, Clauw DJ, Galor A. Antidepressant Medication Use for Treatment of Chronic Ocular Pain. Cornea 2024; 43:1335-1339. [PMID: 39348714 PMCID: PMC11444518 DOI: 10.1097/ico.0000000000003646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 06/23/2024] [Indexed: 10/02/2024]
Abstract
ABSTRACT Chronic ocular pain (COP) is a leading cause of eye care visits in the US and has a substantial impact on quality of life and visual functioning. Although many conditions underlie COP, such as dry eye disease or post-herpetic neuralgia, some people experience pain without significant ocular signs on examination or known risk factors (eg, traumatic injury). Antidepressant medications that act primarily on the central nervous system, such as tricyclic antidepressants and serotonin and norepinephrine reuptake inhibitors, are often used to treat patients with COP, but a recent Cochrane review and network meta-analysis investigating 25 different antidepressants concluded that only two serotonin and norepinephrine reuptake inhibitors, have sufficient data to support their use in chronic pain. For all other medications, the evidence was of low certainty. We contend that while these medications are not a cure-all for chronic pain or COP, it is premature to conclude that these medications have no role in their treatment. We provide a rationale for continued use of antidepressant medications as part of a multimodal targeted treatment for patients with COP.
Collapse
Affiliation(s)
- Lindsey B De Lott
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, MI
| | - Noreen Khan
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, MI
| | - Maria A Woodward
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, MI
| | - Daniel J Clauw
- Department of Anesthesiology, Chronic Pain and Fatigue Research Center University of Michigan Medical School, Ann Arbor, MI; and
| | - Anat Galor
- Surgical Services, Miami Veterans Administration Medical Center, Bascom Palmer Eye Institute, University of Miami, Miami, FL
| |
Collapse
|
5
|
Chelucci E, Daniele S, Vergassola M, Ceccarelli L, Zucchi S, Boltri L, Martini C. Trazodone counteracts the response of microglial cells to inflammatory stimuli. Eur J Neurosci 2024; 60:5605-5620. [PMID: 39187397 DOI: 10.1111/ejn.16522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 07/29/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024]
Abstract
Microglia are resident brain cells that regulate neuronal development and innate immunity. Microglia activation participates in the cellular response to neuroinflammation, thus representing a possible target for pharmacological strategies aimed to counteract the onset and progression of brain disorders, including depression. Antidepressant drugs have been reported to reduce neuroinflammation by acting also on glial cells. Herein, the potential anti-inflammatory and neuroprotective effects of trazodone (TRZ) on the microglial human microglial clone 3 (HMC3) cell line were investigated. HMC3 cells were activated by a double inflammatory stimulus (lipopolysaccharide [LPS] and tumour necrosis factor-alpha [TNF-α], 24 h each), and the induction of inflammation was demonstrated by (i) the increased expression levels of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) and ionized calcium-binding adapter molecule 1 (IBA-1), and (ii) the increased release of interleukin 6 (IL-6) and transforming growth factor-beta (TGF-β). TRZ effects were evaluated by treating HMC3 cells for 24 h before (pre-treatment) and after (post-treatment) the double inflammatory stimulus. Notably, TRZ treatments significantly decreased the expression of NF-kB and IBA-1 and the release of the cytokines IL-6 and TGF-β. Moreover, TRZ prevented and reduced the release of quinolinic acid (QUIN), a known neurotoxic kynurenine metabolite. Finally, cellular supernatants collected from microglial cells pre-treated LPS-TNF-α with TRZ were able to improve neuronal-like cell viability, demonstrating a potential neuroprotective effect. Overall, this study suggests the anti-inflammatory effects of TRZ on human microglia and strives for its neuroprotective properties.
Collapse
Affiliation(s)
| | | | - Matteo Vergassola
- Angelini Pharma S.p.A. Global External Innovation & Drug Discovery, Translational Research Department, Rome, Italy
| | | | - Sara Zucchi
- Angelini Pharma S.p.A. Global R&D PLCM Preclinical Development, Ancona, Italy
| | - Luigi Boltri
- Angelini Pharma S.p.A. Global R&D PLCM Preclinical Development, Ancona, Italy
| | | |
Collapse
|
6
|
Comanescu C, Racovita RC. An Overview of Degradation Strategies for Amitriptyline. Int J Mol Sci 2024; 25:3822. [PMID: 38612638 PMCID: PMC11012176 DOI: 10.3390/ijms25073822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 03/12/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Antidepressant drugs play a crucial role in the treatment of mental health disorders, but their efficacy and safety can be compromised by drug degradation. Recent reports point to several drugs found in concentrations ranging from the limit of detection (LOD) to hundreds of ng/L in wastewater plants around the globe; hence, antidepressants can be considered emerging pollutants with potential consequences for human health and wellbeing. Understanding and implementing effective degradation strategies are essential not only to ensure the stability and potency of these medications but also for their safe disposal in line with current environment remediation goals. This review provides an overview of degradation pathways for amitriptyline, a typical tricyclic antidepressant drug, by exploring chemical routes such as oxidation, hydrolysis, and photodegradation. Connex issues such as stability-enhancing approaches through formulation and packaging considerations, regulatory guidelines, and quality control measures are also briefly noted. Specific case studies of amitriptyline degradation pathways forecast the future perspectives and challenges in this field, helping researchers and pharmaceutical manufacturers to provide guidelines for the most effective degradation pathways employed for minimal environmental impact.
Collapse
Affiliation(s)
- Cezar Comanescu
- Department of Inorganic Chemistry, Physical Chemistry and Electrochemistry, Faculty of Chemical Engineering and Biotechnologies, National University of Science and Technology POLITEHNICA Bucharest, 1-7 Gh. Polizu St., District 1, 011061 Bucharest, Romania
- National Institute of Materials Physics, Atomistilor 405A, 077125 Magurele, Romania
- Faculty of Physics, University of Bucharest, Atomistilor 405, 077125 Magurele, Romania
| | - Radu C. Racovita
- Department of Inorganic Chemistry, Physical Chemistry and Electrochemistry, Faculty of Chemical Engineering and Biotechnologies, National University of Science and Technology POLITEHNICA Bucharest, 1-7 Gh. Polizu St., District 1, 011061 Bucharest, Romania
| |
Collapse
|
7
|
Santandreu J, Caballero FF, Gómez-Serranillos MP, González-Burgos E. Risk of dementia among antidepressant elderly users: A population-based cohort analysis in Spain. J Affect Disord 2024; 349:54-61. [PMID: 38195007 DOI: 10.1016/j.jad.2024.01.002] [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: 07/26/2023] [Revised: 11/21/2023] [Accepted: 01/03/2024] [Indexed: 01/11/2024]
Abstract
The use of antidepressants with anticholinergic effects has been associated with an increased risk of dementia. However, the results published are contradictory. The aim of the study is to compare the risk of developing dementia in elderly who were prescribed tricyclic antidepressants (TCA) versus those who were prescribed selective serotonin reuptake inhibitors (SSRIs) and other antidepressants (OA). A prospective population-based cohort study was performed using the Spanish Database for Pharmacoepidemiological Research in Primary Care (BIFAP) data (from 2005 to 2018). The cohort study included 62,928 patients age ≥ 60 without dementia and with antidepressant long-term monotherapy. Patients were divided into exposure antidepressant groups based on ATC system [TCA, SSRIs users and OAs users]. The risk of dementia was calculated by Cox regression models, providing hazard ratios (HR) and 95 % confidence intervals. The Kaplan-Meier model was used for survival analysis. Chi2 test was used as association test. The results showed SSRI users had higher dementia risk than TCA users (HR = 1.864; 95%CI = 1.624-2.140). Moreover, OA users had also significant risk of dementia (HR = 2.103; 95%CI = 1.818-2.431). Several limitations are the variation of the trend in the prescription of antidepressants, the small number of patients that use some antidepressants, the lack of information related to the dose, or socioeconomic characteristics, the use of antidepressant drugs for other indications, or the therapeutic compliance. Our findings showed that older users of SSRI and OA have more risk of developing dementia than TCA elderly users. However, additional studies would be needed.
Collapse
Affiliation(s)
- Javier Santandreu
- Department of Pharmacology, Pharmacognosy and Botany, Faculty of Pharmacy, Universidad Complutense de Madrid, Madrid, Spain
| | - Francisco Félix Caballero
- Department of Preventive Medicine and Public Health, School of Medicine, Universidad Autónoma de Madrid and CIBERESP (CIBER of Epidemiology and Public Health), Madrid, Spain
| | - M Pilar Gómez-Serranillos
- Department of Pharmacology, Pharmacognosy and Botany, Faculty of Pharmacy, Universidad Complutense de Madrid, Madrid, Spain
| | - Elena González-Burgos
- Department of Pharmacology, Pharmacognosy and Botany, Faculty of Pharmacy, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
8
|
Cobos-Campos R, de Lafuente-Moríñigo AS, Cordero-Guevara JA, Bermúdez-Ampudia C, Apiñaniz A, Parraza N. Effectiveness of antidepressants in improving the prognosis of COVID-19: A systematic review and meta-analysis. Aten Primaria 2024; 56:102771. [PMID: 38016405 PMCID: PMC10696393 DOI: 10.1016/j.aprim.2023.102771] [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: 06/23/2023] [Revised: 08/04/2023] [Accepted: 08/25/2023] [Indexed: 11/30/2023] Open
Abstract
OBJECTIVE, AND MATERIAL AND METHODS A systematic review and meta-analysis was performed to evaluate the effectiveness of antidepressants in reducing the poor evolution of COVID-19 disease (a composite variable including death, hospitalization and need for mechanical ventilation), and mortality, according the guidelines for Systematic Reviews of Interventions published by the Cochrane library. SOURCE OF DATA MEDLINE, EMBASE and COCHRANE LIBRARY were consulted up to February 25, 2022. Unpublished studies were searched on clinicaltrials.gov platform. SELECTION OF STUDIES Seven masked and unmasked, observational and experimental studies evaluating death, hospitalization and need for mechanical ventilation were selected. A second subgroup analysis with mortality variable was performed. DATA EXTRACTION A full risk of bias assessment was performed addressing issues such as information and confounding bias. ROB2 and Robins-I tools for randomized and no randomized studies were employed respectively. In the quantitative analysis, the risk of publication bias, heterogeneity, estimation of pooled measure and a sensitivity analysis was performed. The pooled final measure was calculated as odds ratio with its correspondent 95% confidence interval. A random effects model was used for this purpose due to the heterogeneity between included studies. Finally, a sensitivity analysis was performed to assess the robustness of final pooled measure. RESULTS Seven studies were finally considered to calculate the final pooled measure. The effect of intervention was OR 0.73; 95% CI 0.56-0.94. CONCLUSIONS The use of antidepressants, and specially SSRI could be effective for reducing the risk of poor progression of COVID-19 disease.
Collapse
Affiliation(s)
- Raquel Cobos-Campos
- Bioaraba Health Research Institute, Epidemiology and Public Health Group, Vitoria-Gasteiz, Spain.
| | | | | | | | - Antxon Apiñaniz
- Bioaraba Health Research Institute, Epidemiology and Public Health Group, Vitoria-Gasteiz, Spain; Osakidetza Basque Health Service, Aranbizkarra I Health Center, Vitoria-Gasteiz, Spain; Department of Preventive Medicine and Public Health, University of the Basque Country, Vitoria-Gasteiz, Spain
| | - Naiara Parraza
- Bioaraba Health Research Institute, Epidemiology and Public Health Group, Vitoria-Gasteiz, Spain
| |
Collapse
|
9
|
Rajkumar RP. Resolving a paradox: antidepressants, neuroinflammation, and neurodegeneration. EXPLORATION OF NEUROPROTECTIVE THERAPY 2024:11-37. [DOI: 10.37349/ent.2024.00068] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2025]
Abstract
Depression is a known risk factor for dementia. Antidepressants are the most commonly used treatment for this condition, and are effective in at least half to two-thirds of cases. Extensive evidence from in vitro and animal models suggests that antidepressants have anti-inflammatory and neuroprotective properties. These effects have been shown to reduce the oxidative damage, amyloid aggregation, and expression of pro-inflammatory genes associated with animal models of neurodegenerative disorders. However, longitudinal research in humans has shown that antidepressants do not protect against dementia, and may even be associated with a risk of cognitive deterioration over time in older adults. The contrast between two sets of findings represents a paradox of significant clinical and public health significance, particularly when treating depression in late life. This review paper attempts to resolve this paradox by critically reviewing the medium- and long-term effects of antidepressants on peripheral immune-inflammatory responses, infection risk, gut microbiota, and neuroendocrine responses to stress, and how these effects may influence the risk of neurodegeneration. Briefly stated, it is possible that the peripheral actions of antidepressant medications may antagonize their beneficial effects against neuroinflammation. The implications of these findings are then explored with a particular focus on the development and testing of multimodal neuroprotective and anti-inflammatory treatments that could reduce the risk of Alzheimer’s and related dementias in patients suffering from depression.
Collapse
Affiliation(s)
- Ravi Philip Rajkumar
- Department of Psychiatry, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry 605006, Pondicherry, India
| |
Collapse
|
10
|
Pastis I, Santos MG, Paruchuri A. Exploring the role of inflammation in major depressive disorder: beyond the monoamine hypothesis. Front Behav Neurosci 2024; 17:1282242. [PMID: 38299049 PMCID: PMC10829100 DOI: 10.3389/fnbeh.2023.1282242] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 12/04/2023] [Indexed: 02/02/2024] Open
Abstract
Major depressive disorder affects approximately 8.4% of the United States population. The World Health Organization estimates that 280 million adults worldwide are suffering from depression. They have estimated that by 2030 it will be the second most serious condition. Current treatment relies on the monoamine hypothesis, however, one-third of patients with MDD do not respond to monoamine-based antidepressants. For years, it was hypothesized that the primary pathway of MDD involved serotonin as the main neurotransmitter. The monoamine hypothesis, a widely accepted theory, sought to explain the biological basis of MDD as being caused by the depletion of monoamine neurotransmitters, namely norepinephrine and serotonin. This hypothesis regarding monoamines as the pathophysiological basis of MDD led to the design and widespread use of selective serotonin reuptake inhibitors. However, given that only one-third of patients improve with SSRI it is reasonable to infer that the pathway involved is more complex than once hypothesized and there are more neurotransmitters, receptors, and molecules involved. The monoamine hypothesis does not explain why there is a delay in the onset of effect and action of SSRIs. Several studies have demonstrated that chronic stress is a risk factor for the development of MDD. Thus the monoamine hypothesis alone is not enough to fully account for the pathophysiology of MDD highlighting the need for further research involving the pathways of MDD. In this paper, we review the role of inflammation and cytokines on MDD and discuss other pathways involved in the development and persistence of depressive symptoms.
Collapse
Affiliation(s)
- Irene Pastis
- Department of Psychiatry and Behavioral Medicine, East Carolina University, Greenville, NC, United States
| | - Melody G. Santos
- Internal Medicine and Psychiatry Combined Program, Department of Psychiatry and Behavioral Medicine, East Carolina University, Greenville, NC, United States
| | - Akshita Paruchuri
- East Carolina University Brody School of Medicine, Greenville, NC, United States
| |
Collapse
|
11
|
Hanna DA, Messiha BAS, Abo-Saif AA, Ali FEM, Azouz AA. Lysosomal membrane stabilization by imipramine attenuates gentamicin-induced renal injury: Enhanced LAMP2 expression, down-regulation of cytoplasmic cathepsin D and tBid/cytochrome c/cleaved caspase-3 apoptotic signaling. Int Immunopharmacol 2024; 126:111179. [PMID: 37995569 DOI: 10.1016/j.intimp.2023.111179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 10/13/2023] [Accepted: 11/01/2023] [Indexed: 11/25/2023]
Abstract
Nephrotoxicity is a serious complication commonly encountered with gentamicin (GTM) treatment. Permeabilization of lysosomes with subsequent cytoplasmic release of GTM and cathepsins is considered a crucial issue in progression of GTM toxicity. This study was designed to evaluate the prospective defensive effect of lysosomal membrane stabilization by imipramine (IMP) against GTM nephrotoxicity in rats. GTM (30 mg/kg/h) was intraperitoneally administered over 4 h daily (120 mg/kg/day) for 7 days. IMP (30 mg/kg/day) was orally administered for 14 days; starting 7 days before and then concurrently with GTM. On 15th day, samples (urine, blood, kidney) were collected to estimate biomarkers of kidney function, lysosomal stability, apoptosis, and inflammation. IMP administration to GTM-treated rats ameliorated the disruption in lysosomal membrane stability induced by GTM. That was evidenced by enhanced renal protein expressions of LAMP2 and PI3K, but reduced cathepsin D cytoplasmic expression in kidney sections. Besides, IMP guarded against apoptosis in GTM-treated rats by down-regulation of the pro-apoptotic (tBid, Bax, cytochrome c) and the effector cleaved caspase-3 expressions, while the anti-apoptotic Bcl-2 expression was enhanced. Additionally, the inflammatory cascade p38 MAPK/NF-κB/TNF-α was attenuated in GTM + IMP group along with marked improvement in kidney function biomarkers, compared to GTM group. These findings were supported by the obvious improvement in histological architecture. Furthermore, in vitro enhancement of the antibacterial activity of GTM by IMP confers an additional benefit to their combination. Conclusively, lysosomal membrane stabilization by IMP with subsequent suppression of tBid/cytochrome c/cleaved caspase-3 apoptotic signaling could be a promising protective strategy against GTM nephrotoxicity.
Collapse
Affiliation(s)
- Dina A Hanna
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Basim A S Messiha
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Ali A Abo-Saif
- Department of Pharmacology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Fares E M Ali
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Amany A Azouz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt.
| |
Collapse
|
12
|
Sanchez-Ruiz JA, Leibman NI, Larson NB, Jenkins GD, Ahmed AT, Nunez NA, Biernacka JM, Winham SJ, Weinshilboum RM, Wang L, Frye MA, Ozerdem A. Age-Dependent Sex Differences in the Prevalence of Selective Serotonin Reuptake Inhibitor Treatment: A Retrospective Cohort Analysis. J Womens Health (Larchmt) 2023; 32:1229-1240. [PMID: 37856151 PMCID: PMC10621660 DOI: 10.1089/jwh.2022.0484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023] Open
Abstract
Background: Antidepressants are among the most prescribed medications in the United States. The aim of this study was to explore the prevalence of antidepressant prescriptions and investigate sex differences and age-sex interactions in adults enrolled in the Right Drug, Right Dose, Right Time: Using Genomic Data to Individualize Treatment (RIGHT) study. Materials and Methods: We conducted a retrospective analysis of the RIGHT study. Using electronic prescriptions, we assessed 12-month prevalence of antidepressant treatment. Sex differences and age-sex interactions were evaluated using multivariable logistic regression and flexible recursive smoothing splines. Results: The sample consisted of 11,087 participants (60% women). Antidepressant prescription prevalence was 22.24% (27.96% women, 13.58% men). After adjusting for age and enrollment year, women had significantly greater odds of antidepressant prescription (odds ratio = 2.29; 95% confidence interval = 2.07, 2.54). Furthermore, selective serotonin reuptake inhibitors (SSRIs) had a significant age-sex interaction. While SSRI prescriptions in men showed a sustained decrease with age, there was no such decline for women until after reaching ∼50 years of age. There are important limitations to consider in this study. Electronic prescription data were cross-sectional; information on treatment duration or adherence was not collected; this cohort is not nationally representative; and enrollment occurred over a broad period, introducing confounding by changes in temporal prescribing practices. Conclusions: Underscored by the significant interaction between age and sex on odds of SSRI prescription, our results warrant age to be incorporated as a mediator when investigating sex differences in mental illness, especially mood disorders and their treatment.
Collapse
Affiliation(s)
| | - Nicole I. Leibman
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Nicholas B. Larson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Gregory D. Jenkins
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Ahmed T. Ahmed
- The Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA
| | - Nicolas A. Nunez
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, Minnesota, USA
| | - Joanna M. Biernacka
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Stacey J. Winham
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Richard M. Weinshilboum
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Liewei Wang
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Mark A. Frye
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, Minnesota, USA
| | - Aysegul Ozerdem
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
13
|
Fang S, Wu Z, Guo Y, Zhu W, Wan C, Yuan N, Chen J, Hao W, Mo X, Guo X, Fan L, Li X, Chen J. Roles of microglia in adult hippocampal neurogenesis in depression and their therapeutics. Front Immunol 2023; 14:1193053. [PMID: 37881439 PMCID: PMC10597707 DOI: 10.3389/fimmu.2023.1193053] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/20/2023] [Indexed: 10/27/2023] Open
Abstract
Adult hippocampal neurogenesis generates functional neurons from neural progenitor cells in the hippocampal dentate gyrus (DG) to complement and repair neurons and neural circuits, thus benefiting the treatment of depression. Increasing evidence has shown that aberrant microglial activity can disrupt the appropriate formation and development of functional properties of neurogenesis, which will play a crucial role in the occurrence and development of depression. However, the mechanisms of the crosstalk between microglia and adult hippocampal neurogenesis in depression are not yet fully understood. Therefore, in this review, we first introduce recent discoveries regarding the roles of microglia and adult hippocampal neurogenesis in the etiology of depression. Then, we systematically discuss the possible mechanisms of how microglia regulate adult hippocampal neurogenesis in depression according to recent studies, which involve toll-like receptors, microglial polarization, fractalkine-C-X3-C motif chemokine receptor 1, hypothalamic-pituitary-adrenal axis, cytokines, brain-derived neurotrophic factor, and the microbiota-gut-brain axis, etc. In addition, we summarize the promising drugs that could improve the adult hippocampal neurogenesis by regulating the microglia. These findings will help us understand the complicated pathological mechanisms of depression and shed light on the development of new treatment strategies for this disease.
Collapse
Affiliation(s)
- Shaoyi Fang
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Zhibin Wu
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Yali Guo
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Wenjun Zhu
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Chunmiao Wan
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Naijun Yuan
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
- Shenzhen People’s Hospital, 2Clinical Medical College, Jinan University, Shenzhen, China
| | - Jianbei Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wenzhi Hao
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaowei Mo
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaofang Guo
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Lili Fan
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaojuan Li
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Jiaxu Chen
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
14
|
Cai Y, Zhu ZH, Li RH, Yin XY, Chen RF, Man LJ, Hou WL, Zhu HL, Wang J, Zhang H, Jia QF, Hui L. Association between increased serum interleukin-8 levels and improved cognition in major depressive patients with SSRIs. BMC Psychiatry 2023; 23:122. [PMID: 36823619 PMCID: PMC9948487 DOI: 10.1186/s12888-023-04616-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/19/2023] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND The effect of neuroinflammatory cytokines on cognitive deficits in patients with major depressive disorder (MDD) can be altered by selective serotonin reuptake inhibitors (SSRIs). This study aimed to examine serum interleukin-8 (IL-8) levels, cognitive function, and their associations in MDD patients with SSRIs. METHODS Thirty SSRI-treated MDD patients and 101 healthy controls were recruited for this study. We examined cognitive performance using the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) and serum IL-8 levels using the Human Inflammatory Cytokine Cytometric Bead Array in both cases and controls. RESULTS The RBANS test scores were significantly lower in MDD patients with SSRIs than in healthy controls after controlling for covariates (all p < 0.001). Serum levels of IL-8 were higher in MDD patients with SSRIs than in healthy controls after adjusting for covariates (F = 3.82, p = 0.05). Serum IL-8 levels were positively correlated with sub-scores of delayed memory (r = 0.37, p = 0.04) and visuospatial/constructional (r = 0.43, p = 0.02) in MDD patients with SSRIs but not in in healthy controls (delayed memory score: r = -0.12, p = 0.24; visuospatial/constructional score: r = 0.02, p = 0.81). CONCLUSIONS Our findings suggested that increased serum IL-8 level might not only be involved in the MDD psychopathology or the use of SSRIs but also correspond to improving MDD delayed memory and visuospatial/constructional function.
Collapse
Affiliation(s)
- Yuan Cai
- grid.268099.c0000 0001 0348 3990School of Mental Health, Wenzhou Medical University, Wenzhou, 325035 Zhejiang People’s Republic of China ,grid.263761.70000 0001 0198 0694Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, No. 11 Guangqian Road, Suzhou, 215137 Jiangsu People’s Republic of China
| | - Zhen Hua Zhu
- grid.263761.70000 0001 0198 0694Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, No. 11 Guangqian Road, Suzhou, 215137 Jiangsu People’s Republic of China
| | - Rong Hua Li
- grid.263761.70000 0001 0198 0694Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, No. 11 Guangqian Road, Suzhou, 215137 Jiangsu People’s Republic of China
| | - Xu Yuan Yin
- grid.263761.70000 0001 0198 0694Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, No. 11 Guangqian Road, Suzhou, 215137 Jiangsu People’s Republic of China
| | - Ru Feng Chen
- grid.263761.70000 0001 0198 0694Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, No. 11 Guangqian Road, Suzhou, 215137 Jiangsu People’s Republic of China
| | - Li Juan Man
- grid.263761.70000 0001 0198 0694Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, No. 11 Guangqian Road, Suzhou, 215137 Jiangsu People’s Republic of China
| | - Wen Long Hou
- grid.263761.70000 0001 0198 0694Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, No. 11 Guangqian Road, Suzhou, 215137 Jiangsu People’s Republic of China
| | - Hong Liang Zhu
- grid.263761.70000 0001 0198 0694Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, No. 11 Guangqian Road, Suzhou, 215137 Jiangsu People’s Republic of China
| | - Jing Wang
- grid.263761.70000 0001 0198 0694Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, No. 11 Guangqian Road, Suzhou, 215137 Jiangsu People’s Republic of China
| | - Huiping Zhang
- grid.189504.10000 0004 1936 7558Departments of Psychiatry and Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118-2526 USA
| | - Qiu Fang Jia
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, No. 11 Guangqian Road, Suzhou, 215137, Jiangsu, People's Republic of China.
| | - Li Hui
- School of Mental Health, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China. .,Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, No. 11 Guangqian Road, Suzhou, 215137, Jiangsu, People's Republic of China.
| |
Collapse
|
15
|
Hosseinalizadeh H, Ebrahimi A, Tavakoli A, Monavari SH. Glioblastoma as a Novel Drug Repositioning Target: Updated State. Anticancer Agents Med Chem 2023; 23:1253-1264. [PMID: 36733195 DOI: 10.2174/1871520623666230202163112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/20/2022] [Accepted: 11/22/2022] [Indexed: 02/04/2023]
Abstract
Glioblastoma multiforme (GBM) is an aggressive form of adult brain tumor that can arise from a low-grade astrocytoma. In recent decades, several new conventional therapies have been developed that have significantly improved the prognosis of patients with GBM. Nevertheless, most patients have a limited long-term response to these treatments and survive < 1 year. Therefore, innovative anti-cancer drugs that can be rapidly approved for patient use are urgently needed. One way to achieve accelerated approval is drug repositioning, extending the use of existing drugs for new therapeutic purposes, as it takes less time to validate their biological activity as well as their safety in preclinical models. In this review, a comprehensive analysis of the literature search was performed to list drugs with antiviral, antiparasitic, and antidepressant properties that may be effective in GBM and their putative anti-tumor mechanisms in GBM cells.
Collapse
Affiliation(s)
- Hamed Hosseinalizadeh
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ammar Ebrahimi
- Department of Biomedical Sciences, University of Lausanne, Rue Du Bugnon, Lausanne, Switzerland
| | - Ahmad Tavakoli
- Research Center of Pediatric Infectious Diseases, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
16
|
Antidepressants as a potential candidate to reduce microglia activation in neurodegenerative diseases. A systematic review and meta-analysis of preclinical studies. JOURNAL OF AFFECTIVE DISORDERS REPORTS 2023. [DOI: 10.1016/j.jadr.2023.100465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
17
|
Acid Sphingomyelinase Inhibitor, Imipramine, Reduces Hippocampal Neuronal Death after Traumatic Brain Injury. Int J Mol Sci 2022; 23:ijms232314749. [PMID: 36499076 PMCID: PMC9740309 DOI: 10.3390/ijms232314749] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Traumatic brain injury (TBI) broadly degrades the normal function of the brain after a bump, blow, or jolt to the head. TBI leads to the aggravation of pre-existing brain dysfunction and promotes neurotoxic cascades that involve processes such as oxidative stress, loss of dendritic arborization, and zinc accumulation. Acid sphingomyelinase (ASMase) is an enzyme that hydrolyzes sphingomyelin to ceramide in cells. Under normal conditions, ceramide plays an important role in various physiological functions, such as differentiation and apoptosis. However, under pathological conditions, excessive ceramide production is toxic and activates the neuronal-death pathway. Therefore, we hypothesized that the inhibition of ASMase activity by imipramine would reduce ceramide formation and thus prevent TBI-induced neuronal death. To test our hypothesis, an ASMase inhibitor, imipramine (10 mg/kg, i.p.), was administrated to rats immediately after TBI. Based on the results of this study, we confirmed that imipramine significantly reduced ceramide formation, dendritic loss, oxidative stress, and neuronal death in the TBI-imipramine group compared with the TBI-vehicle group. Additionally, we validated that imipramine prevented TBI-induced cognitive dysfunction and the modified neurological severity score. Consequently, we suggest that ASMase inhibition may be a promising therapeutic strategy to reduce hippocampal neuronal death after TBI.
Collapse
|
18
|
Hatamnejad MR, Baradaran Ghavami S, Shirvani M, Asghari Ahmadabad M, Shahrokh S, Farmani M, Sherkat G, Asadzadeh Aghdaei H, Zali MR. Selective serotonin reuptake inhibitors and inflammatory bowel disease; Beneficial or malpractice. Front Immunol 2022; 13:980189. [PMID: 36275739 PMCID: PMC9583131 DOI: 10.3389/fimmu.2022.980189] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/22/2022] [Indexed: 11/21/2022] Open
Abstract
IBD, a chronic inflammatory disease, has been manifested as a growing health problem. No Crohn's and Colitis councils have officially ratified anti-depressants as a routine regimen for IBD patients. However, some physicians empirically prescribe them to rectify functional bowel consequences such as pain and alleviate psychiatric comorbidities. On the other side, SSRIs' prescription is accompanied by adverse effects such as sleep disturbances. Prolonged intermittent hypoxia throughout sleep disturbance such as sleep apnea provokes periodic reductions in the partial oxygen pressure gradient in the gut lumen. It promotes gut microbiota to dysbiosis, which induces intestinal inflammation. This phenomenon and evidence representing the higher amount of serotonin associated with Crohn's disease challenged our previous knowledge. Can SSRIs worsen the IBD course? Evidence answered the question with the claim on anti-inflammatory properties (central and peripheral) of SSRIs and illuminated the other substantial elements (compared to serotonin elevation) responsible for IBD pathogenesis. However, later clinical evidence was not all in favor of the benefits of SSRIs. Hence, in this review, the molecular mechanisms and clinical evidence are scrutinized and integrated to clarify the interfering molecular mechanism justifying both supporting and disproving clinical evidence. Biphasic dose-dependent serotonin behavior accompanying SSRI shifting function when used up for the long-term can be assumed as the parameters leading to IBD patients' adverse outcomes. Despite more research being needed to elucidate the effect of SSRI consumption in IBD patients, periodic prescriptions of SSRIs at monthly intervals can be recommended.
Collapse
Affiliation(s)
- Mohammad Reza Hatamnejad
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shaghayegh Baradaran Ghavami
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marzieh Shirvani
- Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Shabnam Shahrokh
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Farmani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghazal Sherkat
- Medicine Faculty of Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Asensi-Cantó A, López-Abellán MD, Castillo-Guardiola V, Hurtado AM, Martínez-Penella M, Luengo-Gil G, Conesa-Zamora P. Antitumoral Effects of Tricyclic Antidepressants: Beyond Neuropathic Pain Treatment. Cancers (Basel) 2022; 14:cancers14133248. [PMID: 35805019 PMCID: PMC9265090 DOI: 10.3390/cancers14133248] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/26/2022] [Accepted: 06/28/2022] [Indexed: 11/25/2022] Open
Abstract
Simple Summary Tricyclic antidepressants (TCAs) are old and known therapeutic agents whose good safety profile makes them good candidates for drug repurposing. As the relevance of nerves in cancer development and progression is being unveiled, attention now turns to the use of nerve-targeting drugs, such as TCAs, as an interesting approach to combat cancer. In this review, we discuss current evidence about the safety of TCAs, their application to treat neuropathic pain in cancer patients, and in vitro and in vivo demonstrations of the antitumoral effects of TCAs. Finally, the results of ongoing clinical trials and future directions are discussed. Abstract Growing evidence shows that nerves play an active role in cancer development and progression by altering crucial molecular pathways and cell functions. Conversely, the use of neurotropic drugs, such as tricyclic antidepressants (TCAs), may modulate these molecular signals with a therapeutic purpose based on a direct antitumoral effect and beyond the TCA use to treat neuropathic pain in oncology patients. In this review, we discuss the TCAs’ safety and their central effects against neuropathic pain in cancer, and the antitumoral effects of TCAs in in vitro and preclinical studies, as well as in the clinical setting. The current evidence points out that TCAs are safe and beneficial to treat neuropathic pain associated with cancer and chemotherapy, and they block different molecular pathways used by cancer cells from different locations for tumor growth and promotion. Likewise, ongoing clinical trials evaluating the antineoplastic effects of TCAs are discussed. TCAs are very biologically active compounds, and their repurposing as antitumoral drugs is a promising and straightforward approach to treat specific cancer subtypes and to further define their molecular targets, as well as an interesting starting point to design analogues with increased antitumoral activity.
Collapse
Affiliation(s)
- Antonio Asensi-Cantó
- Facultad de Ciencias de la Salud, Universidad Católica de Murcia (UCAM), 30107 Guadalupe, Spain; (A.A.-C.); (M.D.L.-A.); (M.M.-P.)
- Servicio de Farmacia Hospitalaria, Hospital Universitario Santa Lucía, 30202 Cartagena, Spain
- Grupo de Investigación en Patología Molecular y Farmacogenética, Servicios de Anatomía Patológica y Análisis Clínicos, Instituto Murciano de Investigación Biosanitaria (IMIB), Hospital Universitario Santa Lucía, 30202 Cartagena, Spain; (V.C.-G.); (A.M.H.)
| | - María Dolores López-Abellán
- Facultad de Ciencias de la Salud, Universidad Católica de Murcia (UCAM), 30107 Guadalupe, Spain; (A.A.-C.); (M.D.L.-A.); (M.M.-P.)
- Grupo de Investigación en Patología Molecular y Farmacogenética, Servicios de Anatomía Patológica y Análisis Clínicos, Instituto Murciano de Investigación Biosanitaria (IMIB), Hospital Universitario Santa Lucía, 30202 Cartagena, Spain; (V.C.-G.); (A.M.H.)
| | - Verónica Castillo-Guardiola
- Grupo de Investigación en Patología Molecular y Farmacogenética, Servicios de Anatomía Patológica y Análisis Clínicos, Instituto Murciano de Investigación Biosanitaria (IMIB), Hospital Universitario Santa Lucía, 30202 Cartagena, Spain; (V.C.-G.); (A.M.H.)
| | - Ana María Hurtado
- Grupo de Investigación en Patología Molecular y Farmacogenética, Servicios de Anatomía Patológica y Análisis Clínicos, Instituto Murciano de Investigación Biosanitaria (IMIB), Hospital Universitario Santa Lucía, 30202 Cartagena, Spain; (V.C.-G.); (A.M.H.)
- Grupo de Investigación en Inmunobiología para la Acuicultura, Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
| | - Mónica Martínez-Penella
- Facultad de Ciencias de la Salud, Universidad Católica de Murcia (UCAM), 30107 Guadalupe, Spain; (A.A.-C.); (M.D.L.-A.); (M.M.-P.)
- Servicio de Farmacia Hospitalaria, Hospital Universitario Santa Lucía, 30202 Cartagena, Spain
| | - Ginés Luengo-Gil
- Grupo de Investigación en Patología Molecular y Farmacogenética, Servicios de Anatomía Patológica y Análisis Clínicos, Instituto Murciano de Investigación Biosanitaria (IMIB), Hospital Universitario Santa Lucía, 30202 Cartagena, Spain; (V.C.-G.); (A.M.H.)
- Correspondence: (G.L.-G.); (P.C.-Z.); Tel.: +34-968-128-600 (ext. 951615) (G.L.-G. & P.C.-Z.)
| | - Pablo Conesa-Zamora
- Facultad de Ciencias de la Salud, Universidad Católica de Murcia (UCAM), 30107 Guadalupe, Spain; (A.A.-C.); (M.D.L.-A.); (M.M.-P.)
- Grupo de Investigación en Patología Molecular y Farmacogenética, Servicios de Anatomía Patológica y Análisis Clínicos, Instituto Murciano de Investigación Biosanitaria (IMIB), Hospital Universitario Santa Lucía, 30202 Cartagena, Spain; (V.C.-G.); (A.M.H.)
- Correspondence: (G.L.-G.); (P.C.-Z.); Tel.: +34-968-128-600 (ext. 951615) (G.L.-G. & P.C.-Z.)
| |
Collapse
|
20
|
Shahbazi S, Zakerali T. Methylenedioxy Piperamide-Derived Compound D5 Regulates Inflammatory Cytokine Secretion in a Culture of Human Glial Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27113527. [PMID: 35684465 PMCID: PMC9182381 DOI: 10.3390/molecules27113527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/21/2022] [Accepted: 05/27/2022] [Indexed: 02/07/2023]
Abstract
Neuroinflammation is the cornerstone of most neuronal disorders, particularly neurodegenerative diseases. During the inflammatory process, various pro-inflammatory cytokines, chemokines, and enzymes—such as interleukin 1-β (IL1-β), tumor necrosis factor-α (TNF-α), interleukin 6 (IL-6), inducible nitric oxide synthases (iNOS), inhibitory kappa kinase (IKK), and inducible nitric oxide (NO)—are over-expressed in response to every stimulus. Methods: In the present study, we focused on the anti-neuroinflammatory efficacy of (2E,4E)-N,5-bis(benzo[d][1,3]dioxol-5-yl)penta-2,4-dienamide, encoded D5. We investigated the efficacy of D5 on the upstream and downstream products of inflammatory pathways in CHME3 and SVG cell lines corresponding to human microglia and astrocytes, respectively, using various in silico, in vitro, and in situ techniques. Results: The results showed that D5 significantly reduced the level of pro-inflammatory cytokines by up-regulating PPAR-γ expression and suppressing IKK-β, iNOS, NO production, and NF-κB activation in inflamed astrocytes (SVG) and microglia (CHME3) after 24 h of incubation. The data demonstrated remarkably higher efficacy of D5 compared to ASA (Aspirin) in reducing NF-κB-dependent neuroinflammation. Conclusions: We observed that the functional-group alteration had an extreme influence on the levels of druggability and the immunomodulatory properties of two analogs of piperamide, D5, and D4 ((2E,4E)-5-(benzo[d][1,3]dioxol-5-yl)-N-(4-(hydroxymethyl)phenyl)penta-2,4-dienamide)). The present study suggested D5 as a potential anti-neuroinflammatory agent for further in vitro, in vivo, and clinical investigations.
Collapse
Affiliation(s)
- Sajad Shahbazi
- BRAINCITY, Neurobiology Lab, Nencki Institute of Experimental Biology, 02-093 Warszawa, Poland
- Correspondence:
| | - Tara Zakerali
- Nencki Institute of Experimental Biology, 02-093 Warszawa, Poland;
| |
Collapse
|
21
|
Administration of an Acidic Sphingomyelinase (ASMase) Inhibitor, Imipramine, Reduces Hypoglycemia-Induced Hippocampal Neuronal Death. Cells 2022; 11:cells11040667. [PMID: 35203316 PMCID: PMC8869983 DOI: 10.3390/cells11040667] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/31/2022] [Accepted: 02/12/2022] [Indexed: 01/27/2023] Open
Abstract
Severe hypoglycemia (below 35 mg/dL) appears most often in diabetes patients who continuously inject insulin. To rapidly cease the hypoglycemic state in this study, glucose reperfusion was conducted, which can induce a secondary neuronal death cascade following hypoglycemia. Acid sphingomyelinase (ASMase) hydrolyzes sphingomyelin into ceramide and phosphorylcholine. ASMase activity can be influenced by cations, pH, redox, lipids, and other proteins in the cells, and there are many changes in these factors in hypoglycemia. Thus, we expect that ASMase is activated excessively after hypoglycemia. Ceramide is known to cause free radical production, excessive inflammation, calcium dysregulation, and lysosomal injury, resulting in apoptosis and the necrosis of neurons. Imipramine is mainly used in the treatment of depression and certain anxiety disorders, and it is particularly known as an ASMase inhibitor. We hypothesized that imipramine could decrease hippocampal neuronal death by reducing ceramide via the inhibition of ASMase after hypoglycemia. In the present study, we confirmed that the administration of imipramine significantly reduced hypoglycemia-induced neuronal death and improved cognitive function. Therefore, we suggest that imipramine may be a promising therapeutic tool for preventing hypoglycemia-induced neuronal death.
Collapse
|
22
|
Zhu ZH, Song XY, Man LJ, Chen P, Tang Z, Li RH, Ji CF, Dai NB, Liu F, Wang J, Zhang J, Jia QF, Hui L. Comparisons of Serum Interleukin-8 Levels in Major Depressive Patients With Drug-Free Versus SSRIs Versus Healthy Controls. Front Psychiatry 2022; 13:858675. [PMID: 35492731 PMCID: PMC9046727 DOI: 10.3389/fpsyt.2022.858675] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE The interleukin-8 (IL-8) has been reported to play an important role in depression, which might be modulated by the selective serotonin reuptake inhibitors (SSRIs). Thus, the aim of this study was to investigate serum IL-8 levels, depressive symptom, and their associations in drug-free MDD patients, MDD patients with SSRIs, and healthy controls (HCs). METHODS Fifty-seven drug-free MDD patients (male/female = 35/22, mean age: 39.24 years), 30 MDD patients with SSRIs (male/female = 11/19, mean age: 39.73 years), and 101 HCs (male/female = 52/49, mean age: 37.38 years) were recruited in this cross-sectional study. Serum IL-8 levels and depressive symptom were assessed using the Flow Cytometer and Hamilton Depression Scale (HAMD). The analysis of variance was used for the comparison between groups. The relationship between serum log10 IL-8 levels and HAMD score was analyzed by Pearson correlation. RESULTS Serum log10IL-8 levels were lower in all patients than HCs after controlling for covariates (F = 4.86, p = 0.03). There was significant difference in serum Log10IL-8 levels among three groups after controlling for covariates (F = 14.63, p < 0.001). Serum Log10IL-8 levels in drug-free patients were lower compared to HCs (F = 19.38, p < 0.001) or patients with SSRIs (F = 21.89, p < 0.001) after controlling for covariates. However, there was not difference in serum log10IL-8 levels between patients with SSRIs and HCs after controlling for covariates. Moreover, serum Log10IL-8 levels were negatively correlated with HAMD score in all patients (r = -0.37, p = 0.02). Also, serum Log10IL-8 levels were negatively correlated with HAMD score in drug-free patients (r = -0.74, p = 0.01), but not in patients with SSRIs. CONCLUSION Our data supported that the decline in serum IL-8 levels was association with depression. Moreover, the SSRIs might modulate increased serum IL-8 levels of depression.
Collapse
Affiliation(s)
- Zhen Hua Zhu
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, Suzhou, China
| | - Xiao Ying Song
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, Suzhou, China
| | - Li Juan Man
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, Suzhou, China
| | - Peng Chen
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, Suzhou, China
| | - Zhen Tang
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, Suzhou, China
| | - Rong Hua Li
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, Suzhou, China
| | - Cai Fang Ji
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, Suzhou, China
| | - Ning Bin Dai
- Suzhou Center for Disease Prevention and Control, Suzhou, China
| | - Fang Liu
- Suzhou Center for Disease Prevention and Control, Suzhou, China
| | - Jing Wang
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, Suzhou, China
| | - Jianping Zhang
- Department of Psychiatry, Weill Cornell Medical College, Cornell University, New York, NY, United States
| | - Qiu Fang Jia
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, Suzhou, China
| | - Li Hui
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
23
|
Dimoula A, Fotellis D, Aivalioti E, Delialis D, Polissidis A, Patras R, Kokras N, Stamatelopoulos K. Off-Target Effects of Antidepressants on Vascular Function and Structure. Biomedicines 2021; 10:biomedicines10010056. [PMID: 35052735 PMCID: PMC8773150 DOI: 10.3390/biomedicines10010056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 12/22/2022] Open
Abstract
Depression emerges as a risk factor for cardiovascular disease, and it is thought that successful antidepressant treatment may reduce such a risk. Therefore, antidepressant treatment embodies a potential preventive measure to reduce cardiovascular events in patients with depression. Accumulating evidence indicates that antidepressants have off-target effects on vascular dysfunction and in the early stages of atherosclerosis, which form the basis for cardiovascular disease (CVD) pathogenesis. In this context, we performed a thorough review of the evidence pertaining to the effects of different classes of antidepressant medications on hemodynamic and early atherosclerosis markers. The preclinical and clinical evidence reviewed revealed a preponderance of studies assessing selective serotonin reuptake inhibitors (SSRI), whereas other classes of antidepressants are less well-studied. Sufficient evidence supports a beneficial effect of SSRIs on vascular inflammation, endothelial function, arterial stiffening, and possibly delaying carotid atherosclerosis. In clinical studies, dissecting the hypothesized direct beneficial antidepressant effect of SSRIs on endothelial health from the global improvement upon remission of depression has proven to be difficult. However, preclinical studies armed with appropriate control groups provide evidence of molecular mechanisms linked to endothelial function that are indeed modulated by antidepressants. This suggests at least a partial direct action on vascular integrity. Further research on endothelial markers should focus on the effect of antidepressants on treatment responders versus non-responders in order to better ascertain the possible beneficial vascular effects of antidepressants, irrespective of the underlying course of depression.
Collapse
Affiliation(s)
- Anna Dimoula
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 80 Vas. Sofias Str., 11528 Athens, Greece; (A.D.); (D.F.); (E.A.); (D.D.); (R.P.)
| | - Dimitrios Fotellis
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 80 Vas. Sofias Str., 11528 Athens, Greece; (A.D.); (D.F.); (E.A.); (D.D.); (R.P.)
| | - Evmorfia Aivalioti
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 80 Vas. Sofias Str., 11528 Athens, Greece; (A.D.); (D.F.); (E.A.); (D.D.); (R.P.)
| | - Dimitrios Delialis
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 80 Vas. Sofias Str., 11528 Athens, Greece; (A.D.); (D.F.); (E.A.); (D.D.); (R.P.)
| | - Alexia Polissidis
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.P.); (N.K.)
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), 4 Soranou Efesiou St., 11527 Athens, Greece
- First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Raphael Patras
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 80 Vas. Sofias Str., 11528 Athens, Greece; (A.D.); (D.F.); (E.A.); (D.D.); (R.P.)
| | - Nikolaos Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.P.); (N.K.)
- First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 80 Vas. Sofias Str., 11528 Athens, Greece; (A.D.); (D.F.); (E.A.); (D.D.); (R.P.)
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK
- Correspondence:
| |
Collapse
|
24
|
Wang Y, Wang X, Wang X, Wu D, Qi J, Zhang Y, Wang K, Zhou D, Meng QM, Nie E, Wang Q, Yu RT, Zhou XP. Imipramine impedes glioma progression by inhibiting YAP as a Hippo pathway independent manner and synergizes with temozolomide. J Cell Mol Med 2021; 25:9350-9363. [PMID: 34469035 PMCID: PMC8500960 DOI: 10.1111/jcmm.16874] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/06/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
Patients with malignant glioma often suffered from depression, which leads to an increased risk of detrimental outcomes. Imipramine, an FDA‐approved tricyclic antidepressant, has been commonly used to relieve depressive symptoms in the clinic. Recently, imipramine has been reported to participate in the suppression of tumour progression in several human cancers, including prostate cancer, colon cancer and lymphomas. However, the effect of imipramine on malignant glioma is largely unclear. Here, we show that imipramine significantly retarded proliferation of immortalized and primary glioma cells. Mechanistically, imipramine suppressed tumour proliferation by inhibiting yes‐associated protein (YAP), a recognized oncogene in glioma, independent of Hippo pathway. In addition to inhibiting YAP transcription, imipramine also promoted the subcellular translocation of YAP from nucleus into cytoplasm. Consistently, imipramine administration significantly reduced orthotopic tumour progression and prolonged survival of tumour‐bearing mice. Moreover, exogenous overexpression of YAP partially restored the inhibitory effect of imipramine on glioma progression. Most importantly, compared with imipramine or temozolomide (TMZ) monotherapy, combination therapy with imipramine and TMZ exhibited enhanced inhibitory effect on glioma growth both in vitro and in vivo, suggesting the synergism of both agents. In conclusion, we found that tricyclic antidepressant imipramine impedes glioma progression by inhibiting YAP. In addition, combination therapy with imipramine and TMZ may potentially serve as promising anti‐glioma regimens, thus predicting a broad prospect of clinical application.
Collapse
Affiliation(s)
- Yan Wang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiang Wang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Xu Wang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Di Wu
- Pathological Diagnosis Center, Xuzhou Central Hospital, Xuzhou, China
| | - Ji Qi
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Yu Zhang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Kai Wang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Ding Zhou
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Qing-Ming Meng
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Er Nie
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Qiang Wang
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Ru-Tong Yu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiu-Ping Zhou
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
25
|
Snijders GJLJ, Sneeboer MAM, Fernández-Andreu A, Udine E, Boks MP, Ormel PR, van Berlekom AB, van Mierlo HC, Bӧttcher C, Priller J, Raj T, Hol EM, Kahn RS, de Witte LD. Distinct non-inflammatory signature of microglia in post-mortem brain tissue of patients with major depressive disorder. Mol Psychiatry 2021; 26:3336-3349. [PMID: 33028963 DOI: 10.1038/s41380-020-00896-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 08/22/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023]
Abstract
Findings from epidemiological studies, biomarker measurements and animal experiments suggest a role for aberrant immune processes in the pathogenesis of major depressive disorder (MDD). Microglia, the resident immune cells of the brain, are likely to play a key role in these processes. Previous post-mortem studies reported conflicting findings regarding microglial activation and an in-depth profiling of those cells in MDD is lacking. The aim of this study was therefore to characterize the phenotype and function of microglia in MDD. We isolated microglia from post-mortem brain tissue of patients with MDD (n = 13-19) and control donors (n = 12-25). Using flow cytometry and quantitative Polymerase Chain Reaction (qPCR), we measured protein and mRNA levels of a panel of microglial markers across four different brain regions (medial frontal gyrus, superior temporal gyrus, thalamus, and subventricular zone). In MDD cases, we found a significant upregulation of CX3CR1 and TMEM119 mRNA expression and a downregulation of CD163 mRNA expression and CD14 protein expression across the four brain regions. Expression levels of microglial activation markers, such as HLA-DRA, IL6, and IL1β, as well as the inflammatory responses to lipopolysaccharide and dexamethasone were unchanged. Our findings suggest that microglia enhance homeostatic functions in MDD but are not immune activated.
Collapse
Affiliation(s)
- Gijsje J L J Snijders
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.
| | - Marjolein A M Sneeboer
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.,Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands
| | - Alba Fernández-Andreu
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands
| | - Evan Udine
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Marco P Boks
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands
| | - Paul R Ormel
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.,Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands
| | - Amber Berdenis van Berlekom
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.,Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands
| | - Hans C van Mierlo
- Department of Psychiatry, St. Antonius Hospital, Nieuwegein, Koekoekslaan 1, 3430, EM, Nieuwegein, The Netherlands
| | - Chotima Bӧttcher
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Josef Priller
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany.,DZNE and BIH, 10117, Berlin, Germany.,University of Edinburgh and UK DRI, Edinburgh, EH16 4SB, UK
| | - Towfique Raj
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elly M Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.,Neuroimmunology, Netherlands Institute for Neuroscience, an institute of the royal academy of arts and sciences, 1105, BA, Amsterdam, The Netherlands
| | - René S Kahn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.,Mental Illness Research Education Clinical, Centers of Excellence, VA, Mental Health, Veterans, Bronx, NY, USA
| | - Lot D de Witte
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.,Mental Illness Research Education Clinical, Centers of Excellence, VA, Mental Health, Veterans, Bronx, NY, USA
| |
Collapse
|
26
|
Zeng P, Wang XM, Ye CY, Su HF, Fang YY, Zhang T, Tian Q. Mechanistic insights into the anti-depressant effect of emodin: an integrated systems pharmacology study and experimental validation. Aging (Albany NY) 2021; 13:15078-15099. [PMID: 34051074 PMCID: PMC8221295 DOI: 10.18632/aging.203072] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 05/11/2021] [Indexed: 11/25/2022]
Abstract
Depression is a complex neuropsychiatric disease involved multiple targets and signaling pathways. Systems pharmacology studies could potentially present a comprehensive molecular mechanism to delineate the anti-depressant effect of emodin (EMO). In this study, we investigated the anti-depressant effects of EMO in the chronic unpredictable mild stress (CUMS) rat model of depression and gained insights into the underlying mechanisms using systems pharmacology and molecular simulation analysis. Forty-three potential targets of EMO for treatment of depression were obtained. GO biological process analysis suggested that the biological functions of these targets mainly involve the regulation of reactive oxygen species metabolic process, response to lipopolysaccharide, regulation of inflammatory response, etc. KEGG pathway enrichment analysis showed that the PI3K-Akt signaling pathway, insulin resistance, IL-17 signaling pathway were the most significantly enriched signaling pathways. The molecular docking analysis revealed that EMO might have a strong combination with ESR1, AKT1 and GSK3B. Immunohistochemical staining and Western blotting showed that 2 weeks' EMO treatment (80 mg/kg/day) reduced depression related microglial activation, neuroinflammation and altered PI3K-Akt signaling pathway. Our findings provide a systemic pharmacology basis for the anti-depressant effects of EMO.
Collapse
Affiliation(s)
- Peng Zeng
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao-Ming Wang
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chao-Yuan Ye
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hong-Fei Su
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ying-Yan Fang
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi 435000, China
| | - Teng Zhang
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Neurology, Shanxian Central Hospital, The Affiliated Huxi Hospital of Jining Medical College, Heze 274300, China
| | - Qing Tian
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
27
|
Uzzan S, Azab AN. Anti-TNF-α Compounds as a Treatment for Depression. Molecules 2021; 26:molecules26082368. [PMID: 33921721 PMCID: PMC8073844 DOI: 10.3390/molecules26082368] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/14/2021] [Accepted: 04/17/2021] [Indexed: 12/13/2022] Open
Abstract
Millions of people around the world suffer from psychiatric illnesses, causing unbearable burden and immense distress to patients and their families. Accumulating evidence suggests that inflammation may contribute to the pathophysiology of psychiatric disorders such as major depression and bipolar disorder. Copious studies have consistently shown that patients with mood disorders have increased levels of plasma tumor necrosis factor (TNF)-α. Given these findings, selective anti-TNF-α compounds were tested as a potential therapeutic strategy for mood disorders. This mini-review summarizes the results of studies that examined the mood-modulating effects of anti-TNF-α drugs.
Collapse
Affiliation(s)
- Sarit Uzzan
- Department of Clinical Biochemistry and Pharmacology, School for Community Health Professions—Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel;
| | - Abed N. Azab
- Department of Clinical Biochemistry and Pharmacology, School for Community Health Professions—Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel;
- Department of Nursing, School for Community Health Professions—Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel
- Correspondence: ; Tel.: +972-8-6479880; Fax: +972-8-6477683
| |
Collapse
|
28
|
Khushboo, Kumar A, Sharma B. Biomedical Implications of Plant-Based Principles as Antidepressants: Prospects for Novel Drug Development. Mini Rev Med Chem 2021; 22:904-926. [PMID: 33858313 DOI: 10.2174/1389557521666210415112601] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/30/2020] [Accepted: 02/07/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Depression is a most common mental disorder. The symptoms of depression include loss of energy, changes in appetite, more or less sleep, anxiety, low concentration, uncertainty, restlessness, feelings of worthlessness, guilt, or despair, and thoughts of self-harm or suicide. In order to provide safe, efficient and cost-effective medication, the plants based principles in isolation or in combination with traditional antidepressants are attracting increasing attention for depression therapy. METHOD The information regarding the present review and its contents such as collected from published literature materials in different international journals. We have used different search engines such as PubMed, Medline, ResearchGate Google Semantic Scholar and ScienceDirect. For this purpose, the data obtained were properly organized and suitably analyzed to include in this article. RESULTS Most of the phytomolecules isolated from the medicinal plants display antidepressant effect through the synaptic regulation of levels of neurotransmitters such as dopamine, serotonin, and noradrenaline in different parts of the brain. The mechanism of action of phytomolecules also involves negative regulation of the activities of monoamine oxidase (MAO) and acetylcholinesterase (AChE) and prevention of hyperactivity of hypothalamic-pituitary-adrenal (HPA) axis. In addition, the strong antioxidative and antiinflamatory potential of these phytochemicals offer synergy to their antidepressant as well as antipsychosomatic functions. CONCLUSION The application of phytochemicals has proved it to be a safe, cost effective and efficient therapeutic agent to treat the patients suffering from mild to severe state of depression and other psychiatric disorders. The potential phytochemicals may be further optimized using in silico tools to develop better antidepressants and antisychotic agents in future.
Collapse
Affiliation(s)
- Khushboo
- Department of Biochemistry, University of Allahabad. India
| | - Abhishek Kumar
- Department of Biochemistry, University of Allahabad. India
| | - Bechan Sharma
- Department of Biochemistry, University of Allahabad. India
| |
Collapse
|
29
|
Javadpour P, Askari S, Rashidi FS, Dargahi L, Ahmadiani A, Ghasemi R. Imipramine alleviates memory impairment and hippocampal apoptosis in STZ-induced sporadic Alzheimer's rat model: Possible contribution of MAPKs and insulin signaling. Behav Brain Res 2021; 408:113260. [PMID: 33775777 DOI: 10.1016/j.bbr.2021.113260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/11/2021] [Accepted: 03/20/2021] [Indexed: 10/21/2022]
Abstract
Alzheimer's disease (AD) is the most common age-related neurodegenerative disease, associated with several pathophysiological complaints. Impaired insulin signaling in the brain, is one of the important characteristic features of AD which is accompanied by cognitive deficits. According to the multifactorial and complicated pathology of AD, no modifying therapy has been approved yet. Imipramine is a kind of tricyclic antidepressant with reported anti-inflammatory and anti-oxidant effects in the brain. There are controversial studies about the effect of this drug on spatial memory. This study investigates the effect of imipramine on streptozotocin (STZ) induced memory impairment in rats. Pursuing this objective, rats were treated with imipramine 10 or 20 mg/kg i.p. once a day for 14 days. 24 h after the last injection, memory function was evaluated by the Morris water maze (MWM) test in 4 consecutive days. Then, hippocampi were removed and the activity of caspase-3, mitogen activated protein kinases (MAPKs) family and inhibitory phosphorylation of insulin receptor substrate-1 (IRS-1ser307) were analyzed using Western blotting. Results showed that imipramine prevents memory impairment in STZ induced rats and this improvement was accompanied with an increase in ERK activity, reduction of caspase-3 and JNK activity, as well as partial restoration of P38 and IRS-1 activity. In conclusion, our study demonstrated that at least some members of the MAPK family are involved in the neuroprotective effect of imipramine.
Collapse
Affiliation(s)
- Pegah Javadpour
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Askari
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Sadat Rashidi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasoul Ghasemi
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
30
|
Melnikov M, Sviridova A, Rogovskii V, Oleskin A, Boziki M, Bakirtzis C, Kesidou E, Grigoriadis N, Boykо A. Serotoninergic system targeting in multiple sclerosis: the prospective for pathogenetic therapy. Mult Scler Relat Disord 2021; 51:102888. [PMID: 33756440 DOI: 10.1016/j.msard.2021.102888] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/01/2021] [Accepted: 03/05/2021] [Indexed: 02/07/2023]
Abstract
Serotonin (5-hydroxytryptamine) (5-HT) is a neurotransmitter, which mediates neuropsychological functions of the central nervous system (CNS). Recent studies have shown the modulatory effect of 5-HT on gut microbiota functions, which play an essential role in developing CNS inflammatory diseases. Finally, 5-HT is a direct mediator of neuroimmune interaction. The article reviews the literature data on the role of 5-HT in the regulation of neuroinflammation in multiple sclerosis (MS). The influence of 5-HT and selective serotonin reuptake inhibitors (SSRIs) on experimental autoimmune encephalomyelitis (EAE) and MS pathogenesis, as well as the therapeutic potential of serotoninergic drugs as a pathogenetic therapy of MS, are discussed.
Collapse
Affiliation(s)
- Mikhail Melnikov
- Department of Neuroimmunology, Federal Center of Brain research and Neurotechnology of the Federal Medical-Biological Agency of Russia; Department of Neurology, Neurosurgery and Medical Genetics and Department of Molecular Pharmacology and Radiobiology, Pirogov Russian National Research Medical University, Moscow, Russia; Laboratory of Clinical Immunology, National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, Moscow, Russia.
| | - Anastasiya Sviridova
- Department of Neuroimmunology, Federal Center of Brain research and Neurotechnology of the Federal Medical-Biological Agency of Russia; Department of Neurology, Neurosurgery and Medical Genetics and Department of Molecular Pharmacology and Radiobiology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Vladimir Rogovskii
- Department of Neuroimmunology, Federal Center of Brain research and Neurotechnology of the Federal Medical-Biological Agency of Russia; Department of Neurology, Neurosurgery and Medical Genetics and Department of Molecular Pharmacology and Radiobiology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Alexander Oleskin
- General Ecology and Hydrobiology Department, School of Biology, Moscow State University, Moscow, Russia
| | - Marina Boziki
- 2nd Neurological University Department, Aristotle University of Thessaloniki, AHEPA General Hospital, Thessaloniki, Greece
| | - Christos Bakirtzis
- 2nd Neurological University Department, Aristotle University of Thessaloniki, AHEPA General Hospital, Thessaloniki, Greece
| | - Evangelia Kesidou
- 2nd Neurological University Department, Aristotle University of Thessaloniki, AHEPA General Hospital, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- 2nd Neurological University Department, Aristotle University of Thessaloniki, AHEPA General Hospital, Thessaloniki, Greece
| | - Alexey Boykо
- Department of Neuroimmunology, Federal Center of Brain research and Neurotechnology of the Federal Medical-Biological Agency of Russia; Department of Neurology, Neurosurgery and Medical Genetics and Department of Molecular Pharmacology and Radiobiology, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
31
|
Nobile B, Durand M, Olié E, Guillaume S, Molès JP, Haffen E, Courtet P. The Anti-inflammatory Effect of the Tricyclic Antidepressant Clomipramine and Its High Penetration in the Brain Might Be Useful to Prevent the Psychiatric Consequences of SARS-CoV-2 Infection. Front Pharmacol 2021; 12:615695. [PMID: 33767623 PMCID: PMC7985338 DOI: 10.3389/fphar.2021.615695] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
At the time of writing (December 2020), coronavirus disease 2019 (COVID-19) has already caused more than one million deaths worldwide, and therefore, it is imperative to find effective treatments. The “cytokine storm” induced by Severe Acute Respiratory Syndrome-Coronavirus type 2 (SARS-CoV-2) is a good target to prevent disease worsening, as indicated by the results obtained with tocilizumab and dexamethasone. SARS-CoV-2 can also invade the brain and cause neuro-inflammation with dramatic neurological manifestations, such as viral encephalitis. This could lead to potentially incapacitating long-term consequences, such as the development of psychiatric disorders, as previously observed with SARS-CoV. Several pathways/mechanisms could explain the link between viral infection and development of psychiatric diseases, especially neuro-inflammation induced by SARS-CoV-2. Therefore, it is important to find molecules with anti-inflammatory properties that penetrate easily into the brain. For instance, some antidepressants have anti-inflammatory action and pass easily through the blood brain barrier. Among them, clomipramine has shown very strong anti-inflammatory properties in vitro, in vivo (animal models) and human studies, especially in the brain. The aim of this review is to discuss the potential application of clomipramine to prevent post-infectious mental complications. Repositioning and testing antidepressants for COVID-19 management could help to reduce peripheral and especially central inflammation and to prevent the acute and particularly the long-term consequences of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- B Nobile
- Department of Emergency Psychiatry and Acute Care, CHU Montpellier, Montpellier, France.,IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - M Durand
- Pathogenesis and Control of Chronic Infection, University of Montpellier, INSERM, EFS; CHU Montpellier, Montpellier, France
| | - E Olié
- Department of Emergency Psychiatry and Acute Care, CHU Montpellier, Montpellier, France.,IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France.,FondaMental Foundation, Créteil, France
| | - S Guillaume
- Department of Emergency Psychiatry and Acute Care, CHU Montpellier, Montpellier, France.,IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France.,FondaMental Foundation, Créteil, France
| | - J P Molès
- Pathogenesis and Control of Chronic Infection, University of Montpellier, INSERM, EFS; CHU Montpellier, Montpellier, France
| | - E Haffen
- FondaMental Foundation, Créteil, France.,Service de Psychiatrie de l'Adulte, CIC-1431 INSERM, CHU de Besançon, Laboratoire de Neurosciences, Université de Franche-Comté, Besancon, France
| | - P Courtet
- Department of Emergency Psychiatry and Acute Care, CHU Montpellier, Montpellier, France.,IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France.,FondaMental Foundation, Créteil, France
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW The aim of this review was to analyze COVID-19 effect on the biological features of suicidal vulnerability and its interaction with suicide-related biological pathways. We carried out a narrative review of international publications on the interactions of COVID-19 with the biological bases of suicide. RECENT FINDINGS We hypothesize that SARS-CoV-2 interacts with multiple biological processes that underlie suicidal behavior, such as the renin-angiotensin system, nicotinic receptors, and central and systemic inflammation. Social distancing measures may also worsen subjective or objective social disconnection, thus increasing the risk of suicide. Interestingly, the drugs used to prevent suicide could be promising options to counteract brain damage caused by this coronavirus. SARS-CoV-2 interacts with multiple biological pathways involved in suicide and opens a new window for understanding the suicidal process. The development of suicide prevention treatments in the context of a pandemic may benefit from knowledge on these interactions.
Collapse
Affiliation(s)
- I Conejero
- Department of Emergency Psychiatry and Acute Care, Lapeyronie Hospital, CHU Montpellier, Montpellier, France.
- PSNREC, Univ Montpellier, INSERM, CHU de Montpellier, Montpellier, France.
| | - B Nobile
- Department of Emergency Psychiatry and Acute Care, Lapeyronie Hospital, CHU Montpellier, Montpellier, France
- PSNREC, Univ Montpellier, INSERM, CHU de Montpellier, Montpellier, France
| | - E Olié
- Department of Emergency Psychiatry and Acute Care, Lapeyronie Hospital, CHU Montpellier, Montpellier, France
- PSNREC, Univ Montpellier, INSERM, CHU de Montpellier, Montpellier, France
- FondaMental Foundation, Créteil, France
| | - Ph Courtet
- Department of Emergency Psychiatry and Acute Care, Lapeyronie Hospital, CHU Montpellier, Montpellier, France
- PSNREC, Univ Montpellier, INSERM, CHU de Montpellier, Montpellier, France
- FondaMental Foundation, Créteil, France
| |
Collapse
|
33
|
Jansen van Vuren E, Steyn SF, Brink CB, Möller M, Viljoen FP, Harvey BH. The neuropsychiatric manifestations of COVID-19: Interactions with psychiatric illness and pharmacological treatment. Biomed Pharmacother 2021; 135:111200. [PMID: 33421734 PMCID: PMC7834135 DOI: 10.1016/j.biopha.2020.111200] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/15/2020] [Accepted: 12/26/2020] [Indexed: 12/12/2022] Open
Abstract
The recent outbreak of the corona virus disease (COVID-19) has had major global impact. The relationship between severe acute respiratory syndrome coronavirus (SARS-CoV-2) infection and psychiatric diseases is of great concern, with an evident link between corona virus infections and various central and peripheral nervous system manifestations. Unmitigated neuro-inflammation has been noted to underlie not only the severe respiratory complications of the disease but is also present in a range of neuro-psychiatric illnesses. Several neurological and psychiatric disorders are characterized by immune-inflammatory states, while treatments for these disorders have distinct anti-inflammatory properties and effects. With inflammation being a common contributing factor in SARS-CoV-2, as well as psychiatric disorders, treatment of either condition may affect disease progression of the other or alter response to pharmacological treatment. In this review, we elucidate how viral infections could affect pre-existing psychiatric conditions and how pharmacological treatments of these conditions may affect overall progress and outcome in the treatment of SARS-CoV-2. We address whether any treatment-induced benefits and potential adverse effects may ultimately affect the overall treatment approach, considering the underlying dysregulated neuro-inflammatory processes and potential drug interactions. Finally, we suggest adjunctive treatment options for SARS-CoV-2-associated neuro-psychiatric symptoms.
Collapse
Affiliation(s)
- Esmé Jansen van Vuren
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa.
| | - Stephan F Steyn
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Christiaan B Brink
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Marisa Möller
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Francois P Viljoen
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Brian H Harvey
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa; South African MRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
34
|
Hiramoto K, Yamate Y, Matsuda K, Sugiyama D, Iizuka Y. Tranexamic Acid Improves Memory and Learning Abilities in Aging Mice. J Exp Pharmacol 2020; 12:653-663. [PMID: 33376415 PMCID: PMC7755347 DOI: 10.2147/jep.s284532] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/26/2020] [Indexed: 12/21/2022] Open
Abstract
Purpose Although the onset mechanism of Alzheimer’s disease, which co-occurs with aging, has been extensively studied, no effective methods that improve the decline in memory and learning abilities following aging have been developed. Tranexamic acid provided promising results for ameliorating photo-aging and extending the natural lifespan. However, it is unknown whether it affects the decline in memory and learning abilities due to aging. In this study, we examined the effect of tranexamic acid on memory and learning abilities of naturally aging mice. Methods ICR mice were orally administered with tranexamic acid (12 mg/kg/day) three times weekly for 2 years, and their memory and learning abilities were compared between the tranexamic acid-treated and non-treated groups. Results The decline in memory and learning abilities due to aging was ameliorated by tranexamic acid administration. The expression of plasmin and amyloid-β decreased following the treatment with tranexamic acid. Furthermore, the number of M1-type brain macrophages diminished and that of M2 macrophages increased. In addition, administration of tranexamic acid decreased the concentrations of interleukin (IL)-1β and tumor necrosis factor-α, while it increased the levels of IL-10 and transforming growth factor-α in the brain. Conclusion These results indicated that tranexamic acid suppressed the secretion of the inflammatory cytokines aging M1-type macrophages, thereby improving age-related memory and learning abilities.
Collapse
Affiliation(s)
- Keiichi Hiramoto
- Department of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Mie 513-8670, Japan
| | - Yurika Yamate
- Department of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Mie 513-8670, Japan
| | - Kazunari Matsuda
- R&D Department, Daiichi Sankyo Healthcare Co., LTD, Shinagawa-ku, Tokyo 103-8234, Japan
| | - Daijiro Sugiyama
- R&D Department, Daiichi Sankyo Healthcare Co., LTD, Shinagawa-ku, Tokyo 103-8234, Japan
| | - Yasutaka Iizuka
- R&D Department, Daiichi Sankyo Healthcare Co., LTD, Shinagawa-ku, Tokyo 103-8234, Japan
| |
Collapse
|
35
|
Additive Antidepressant Effects of Combined Administration of Ecitalopram and Caloric Restriction in LPS-Induced Neonatal Model of Depression in Rats. ACTA MEDICA BULGARICA 2020. [DOI: 10.2478/amb-2020-0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Background and aims: Increasing evidence indicates that inflammation in the periphery and neuroinflammation in the brain might be involved in the pathophysiology of depressive symptoms in humans. Relatively little is known about the effects of selective serotonin re-uptake inhibitors (SSRI) on individuals exposed to differential dietary regimens, like caloric restriction (CR).
The aim of the current study is to assess the antidepressant and antineuroinflammatory effects of CR in single administration and combined with SSRIsantidepressant escitalopram in LPS-induced model of depression in Wistar rats.
Materials and methods: For this purpose, we used 36 Wistar rats and applied 3 behavioral tests for depression (FST, SPT and NSFT) in animals and an ELISA-method for measurement of brain IL-1beta levels.
Results: Behavioral assessment and results from ELISA-method have shown that CR not only augments the effect of the antidepressant escitalopram on forced swim test (FST) and sucrose preference test (SPT), but also reduces the brain levels of proinflammatory cytokine IL-1beta. Combined with escitalopram, CR enhances antidepressant and antinflamatory properties of this SSRI.
Discussion and conclusion: These results show that the response to antidepressive treatment depends on the diverse dietary regimens, especially low-caloric diet. We suggest that the background of this is augmentation of anidepressant and antineuronflammatory properties of some antidepressants by CR. Manipulation of dietary regimens is attractive and new strategy for the management of pharmacoresistant depression.
Collapse
|
36
|
Shahbazi S, Zakerali T, Frycz BA, Kaur J. The critical role of piperamide derivative D4 in the regulation of inflammatory response by the microglia and astrocytic glial cells. Biomed Pharmacother 2020; 132:110895. [PMID: 33113430 DOI: 10.1016/j.biopha.2020.110895] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
Acute and chronic inflammation in the central nervous system plays a critical role in the development of neurodegenerative disorders. Various pro-inflammatory cytokines, chemokines, and enzymes such as TNF-α, IL1-β, IL-6, COX-1, COX-2, iNOS, IKK, and inducible nitric oxide are expressed in several signalling pathways, and mediate the neuroinflammatory process. ROS and NF-kB nuclear translocation are the two fundamental pathways involved in neuroinflammatory pathogenesis in neuronal and glial cells. In recent years several compoundswere designed to affect the neuroinflammation and suppress neurodegenerative process. Derivatives of natural products (NPs) attract the most attention of drug developers and industries due to their safety and lesser side effects in comparison with generic drugs. One of the most well-known NP is piperine, which is a yellow crystalline alkaloid extracted from black and white pepper. Recently, we developed a novel piperine derivative (((2E,4E)-5-(benzo[d][1,3]dioxol-5-yl)-N-(4-(hydroxymethyl)phenyl)penta-2,4-dienamide, D4) to enhance the specificity and efficacy of the base molecule. Next, we evaluated the potential anti-inflammatory properities of D4 in CHME3 and SVG cell-lines corresponding to human microglia and astrocytes, respectively. Our results indicated that D4 inhibited NF-kB translocation pathway, and significantly reduced transcript and protein levels of pro-inflammatory cytokines in comparison with Aspirin, as a well-known non-selective NSAID. Furthermore, in silico study showed excellent D4 bioavailability in oral administration. The results of the present study suggest a novel molecule with high anti-neuroinflammatory potency for further pre-clinical tests and pharmacological drug investigation.
Collapse
Affiliation(s)
- Sajad Shahbazi
- Nencki Institute of Experimental Biology, BRAINCITY, Warszawa, Poland.
| | - Tara Zakerali
- Nencki Institute of Experimental Biology, BRAINCITY, Warszawa, Poland
| | - Bartosz A Frycz
- Nencki Institute of Experimental Biology, BRAINCITY, Warszawa, Poland
| | - Jagdeep Kaur
- Department of Biotechnology, Panjab University, Chandigarh, India.
| |
Collapse
|
37
|
Jia X, Gao Z, Hu H. Microglia in depression: current perspectives. SCIENCE CHINA-LIFE SCIENCES 2020; 64:911-925. [PMID: 33068286 DOI: 10.1007/s11427-020-1815-6] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/09/2020] [Indexed: 12/11/2022]
Abstract
Major depressive disorder (MDD) is a prevalent psychiatric disease that involves malfunctions of different cell types in the brain. Accumulating studies started to reveal that microglia, the primary resident immune cells, play an important role in the development and progression of depression. Microglia respond to stress-triggered neuroinflammation, and through the release of proinflammatory cytokines and their metabolic products, microglia may modulate the function of neurons and astrocytes to regulate depression. In this review, we focused on the role of microglia in the etiology of depression. We discussed the dynamic states of microglia; the correlative and causal evidence of microglial abnormalities in depression; possible mechanisms of how microglia sense depression-related stress and modulate depression state; and how antidepressive therapies affect microglia. Understanding the role of microglia in depression may shed light on developing new treatment strategies to fight against this devastating mental illness.
Collapse
Affiliation(s)
- Xiaoning Jia
- Department of Psychiatry of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.,The MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, 310012, China
| | - Zhihua Gao
- The MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, 310012, China. .,NHC and CAMS Key Laboratory of Medical Neurobiology, Mental Health Center, Zhejiang University, Hangzhou, 310058, China. .,Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| | - Hailan Hu
- Department of Psychiatry of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China. .,The MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, 310012, China. .,NHC and CAMS Key Laboratory of Medical Neurobiology, Mental Health Center, Zhejiang University, Hangzhou, 310058, China. .,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, 510515, China. .,Fountain-Valley Institute for Life Sciences, Guangzhou, 510530, China. .,Research Units of Brain Mechanisms Underlying Emotion and Emotion disorders, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
38
|
Ota M, Kanie A, Kobayashi Y, Nakajima A, Sato N, Horikoshi M. Pseudo-continuous arterial spin labeling MRI study of patients with obsessive-compulsive disorder. Psychiatry Res Neuroimaging 2020; 303:111124. [PMID: 32563075 DOI: 10.1016/j.pscychresns.2020.111124] [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: 12/12/2019] [Revised: 05/18/2020] [Accepted: 06/11/2020] [Indexed: 10/24/2022]
Abstract
Arterial spin labeling (ASL) magnetic resonance imaging is a novel technique that can measure regional cerebral blood flow (rCBF). Here we used pseudo-continuous ASL (pCASL) to examine the structural and functional imaging data in patients with obsessive-compulsive disorder (OCD). We estimated the gray matter volume imaging and pCASL imaging data by means of a voxel-by-voxel statistical analysis. We evaluated the differences of rCBF and gray matter volume between the OCD patients and healthy subjects. We detected a significant rCBF reduction in OCD patients in the right posterior cingulate extending to the lingual gyrus, thalamus, and hippocampus, and a significant increase in the left temporal gyrus and left frontal white matter region, compared with healthy subjects. We also observed a significant reduction in gray matter volume of OCD patients in the right hippocampus. We also estimated the correlation between the clinical severity of OCD and the rCBF and gray matter volumes, and found significant negative correlations between the severity of illness and the regional gray matter volume in the bilateral anterior cingulate corti. Our study demonstrated significant changes of rCBF in the cortico-striato-thalamo-cortical pathway around the hippocampus in OCD patients. These findings may help to elucidate the pathogenesis of OCD.
Collapse
Affiliation(s)
- Miho Ota
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan; Department of Neuropsychiatry, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8576, Japan.
| | - Ayako Kanie
- National Center for Cognitive Behavior Therapy and Research, National Center of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Yuki Kobayashi
- National Center for Cognitive Behavior Therapy and Research, National Center of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Aiichiro Nakajima
- National Center for Cognitive Behavior Therapy and Research, National Center of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Noriko Sato
- Department of Radiology, National Center of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Masaru Horikoshi
- National Center for Cognitive Behavior Therapy and Research, National Center of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| |
Collapse
|
39
|
Nobile B, Durand M, Olié E, Guillaume S, Molès JP, Haffen E, Courtet P. Clomipramine Could Be Useful in Preventing Neurological Complications of SARS-CoV-2 Infection. J Neuroimmune Pharmacol 2020; 15:347-348. [PMID: 32601885 PMCID: PMC7324075 DOI: 10.1007/s11481-020-09939-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 06/23/2020] [Indexed: 12/20/2022]
Affiliation(s)
- B Nobile
- Department of Emergency Psychiatry and Acute Care, CHU Montpellier, Montpellier, France. .,PSNREC, INSERM, CHU de Montpellier, University Montpellier, Montpellier, France.
| | - M Durand
- Pathogenesis and control of chronic infection, INSERM, EFS; CHU Montpellier, Montpellier, University of Montpellier, Montpellier, France
| | - E Olié
- Department of Emergency Psychiatry and Acute Care, CHU Montpellier, Montpellier, France.,PSNREC, INSERM, CHU de Montpellier, University Montpellier, Montpellier, France.,FondaMental Foundation, Créteil, France
| | - S Guillaume
- Department of Emergency Psychiatry and Acute Care, CHU Montpellier, Montpellier, France.,PSNREC, INSERM, CHU de Montpellier, University Montpellier, Montpellier, France.,FondaMental Foundation, Créteil, France
| | - J P Molès
- Pathogenesis and control of chronic infection, INSERM, EFS; CHU Montpellier, Montpellier, University of Montpellier, Montpellier, France
| | - E Haffen
- FondaMental Foundation, Créteil, France.,Service de Psychiatrie de l'Adulte, CIC-1431 INSERM, CHU de Besançon, Laboratoire de Neurosciences, Université de Franche-Comté, UBFC, Besançon, France
| | - P Courtet
- Department of Emergency Psychiatry and Acute Care, CHU Montpellier, Montpellier, France.,PSNREC, INSERM, CHU de Montpellier, University Montpellier, Montpellier, France.,FondaMental Foundation, Créteil, France
| |
Collapse
|
40
|
Visentin APV, Colombo R, Scotton E, Fracasso DS, da Rosa AR, Branco CS, Salvador M. Targeting Inflammatory-Mitochondrial Response in Major Depression: Current Evidence and Further Challenges. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2972968. [PMID: 32351669 PMCID: PMC7178465 DOI: 10.1155/2020/2972968] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/26/2020] [Accepted: 03/17/2020] [Indexed: 02/07/2023]
Abstract
The prevalence of psychiatric disorders has increased in recent years. Among existing mental disorders, major depressive disorder (MDD) has emerged as one of the leading causes of disability worldwide, affecting individuals throughout their lives. Currently, MDD affects 15% of adults in the Americas. Over the past 50 years, pharmacotherapy, psychotherapy, and brain stimulation have been used to treat MDD. The most common approach is still pharmacotherapy; however, studies show that about 40% of patients are refractory to existing treatments. Although the monoamine hypothesis has been widely accepted as a molecular mechanism to explain the etiology of depression, its relationship with other biochemical phenomena remains only partially understood. This is the case of the link between MDD and inflammation, mitochondrial dysfunction, and oxidative stress. Studies have found that depressive patients usually exhibit altered inflammatory markers, mitochondrial membrane depolarization, oxidized mitochondrial DNA, and thus high levels of both central and peripheral reactive oxygen species (ROS). The effect of antidepressants on these events remains unclear. Nevertheless, the effects of ROS on the brain are well known, including lipid peroxidation of neuronal membranes, accumulation of peroxidation products in neurons, protein and DNA damage, reduced antioxidant defenses, apoptosis induction, and neuroinflammation. Antioxidants such as ascorbic acid, tocopherols, and coenzyme Q have shown promise in some depressive patients, but without consensus on their efficacy. Hence, this paper provides a review of MDD and its association with inflammation, mitochondrial dysfunction, and oxidative stress and is aimed at thoroughly discussing the putative links between these events, which may contribute to the design and development of new therapeutic approaches for patients.
Collapse
Affiliation(s)
| | - Rafael Colombo
- Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, RS 95070 560, Brazil
| | - Ellen Scotton
- Laboratório de Psiquiatria Molecular, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Farmacologia e Terapêutica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Débora Soligo Fracasso
- Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, RS 95070 560, Brazil
| | - Adriane Ribeiro da Rosa
- Laboratório de Psiquiatria Molecular, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Catia Santos Branco
- Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, RS 95070 560, Brazil
| | - Mirian Salvador
- Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, RS 95070 560, Brazil
| |
Collapse
|
41
|
Kodesh A, Sandin S, Reichenberg A, Rotstein A, Pedersen NL, Ericsson M, Karlsson IK, Davidson M, Levine SZ. Antidepressants and the Risk of Dementia: Appropriate Consideration of Confounding by Indication. Am J Geriatr Psychiatry 2020; 28:499-500. [PMID: 31899121 DOI: 10.1016/j.jagp.2019.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 11/26/2022]
Affiliation(s)
- Arad Kodesh
- Department of Community Mental Health, University of Haifa (AK, AR, SZL), Haifa, Israel; Meuhedet Health Services, Mental Health (AK), Tel Aviv, Israel
| | - Sven Sandin
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet (SS, NP, ME, IK), Stockholm, Sweden; The Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai (SS, AR), New York, NY; Department of Psychiatry, Icahn School of Medicine at Mount Sinai (SS, AR), New York, NY
| | - Abraham Reichenberg
- The Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai (SS, AR), New York, NY; Department of Psychiatry, Icahn School of Medicine at Mount Sinai (SS, AR), New York, NY
| | - Anat Rotstein
- Department of Community Mental Health, University of Haifa (AK, AR, SZL), Haifa, Israel
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet (SS, NP, ME, IK), Stockholm, Sweden
| | - Malin Ericsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet (SS, NP, ME, IK), Stockholm, Sweden
| | - Ida K Karlsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet (SS, NP, ME, IK), Stockholm, Sweden; Institute of Gerontology and Aging Research Network - Jönköping (ARN-J), School of Health and Welfare, Jönköping University (IK), Jönköping, Sweden
| | - Michael Davidson
- Sackler Medical School, Tel Aviv University (MD), Tel Aviv, Israel; Nicosia Medical School (MD), Nicosia, Cyprus
| | - Stephen Z Levine
- Department of Community Mental Health, University of Haifa (AK, AR, SZL), Haifa, Israel.
| |
Collapse
|
42
|
Obuchowicz E, Bielecka-Wajdman A, Zieliński M, Machnik G, Gołyszny M, Ludyga T. Imipramine and Venlafaxine Differentially Affect Primary Glial Cultures of Prenatally Stressed Rats. Front Pharmacol 2020; 10:1687. [PMID: 32076407 PMCID: PMC7006619 DOI: 10.3389/fphar.2019.01687] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/24/2019] [Indexed: 12/25/2022] Open
Abstract
Here, we examine the effects of prenatal administration of two antidepressants—imipramine (IMI) and venlafaxine (VEN)—on morphology and activity of a primary glial culture. Microglia are targeted by antidepressants used for antenatal depression and are important regulators of central nervous system development. In this study, female Wistar rats were assigned to one of four groups: a control group that received water ad libitum (1), and groups that received additionally once daily either water (2), IMI (10 mg/kg) (3), or VEN (20 mg/kg) (4) by oral gavage from gestation day 7 to 22. Oral gavage administration induced prenatal stress. Cell cultures were obtained from the brains of 1-day-old pups. Prenatal stress caused a disturbance of sensorimotor function in pups. Prenatal stress also produced alterations in the glial cultures, specifically, an increased percentage of microglia in the mixed glial cultures and an increased percentage of dead cells. Moreover, increased levels of IL1-β, TNF-α, NO, and an increased expression of CX3CR1 mRNA were found in microglia. However, the ratio of Bax/Bcl2 mRNA was reduced. Prenatal stress increased the vulnerability of microglia to lipopolysaccharide (LPS). The mixed glial culture derived from pups exposed to IMI showed greater morphological changes and the highest percentage of microglia. Microglia were characterized by the largest increase in the production of pro-inflammatory cytokines and NO, and the greatest reduction in the expression of CX3CR1 mRNA. Exposure to IMI reduced the effects of LPS on IL-1β production and Bax/Bcl2 mRNA, and exacerbated the effects of LPS on CX3CR1 mRNA expression. Prenatal administration of VEN induced protective effects on microglia, as measured by all studied parameters. Taken together, our data suggest that, by disturbing microglia function, exposure to even mild forms of chronic prenatal stress may predispose individuals to psychiatric or neurodevelopmental disorders. These data also indicate that chronic mild stress sensitizes microglia to immune challenges, which may lead to enhanced neuronal damage in the embryonic brain. The observed detrimental effects of IMI on microglial activity under conditions of prenatal stress may help to explain the teratogenic effects of IMI reported in the literature.
Collapse
Affiliation(s)
- Ewa Obuchowicz
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Anna Bielecka-Wajdman
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Michał Zieliński
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Grzegorz Machnik
- Department of Internal Medicine and Clinical Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Miłosz Gołyszny
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Tomasz Ludyga
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| |
Collapse
|
43
|
Zhang X, Zhu LB, He JH, Zhang HQ, Ji SY, Zhang CN, Hou NN, Huang CP, Zhu JH. Paroxetine suppresses reactive microglia-mediated but not lipopolysaccharide-induced inflammatory responses in primary astrocytes. J Neuroinflammation 2020; 17:50. [PMID: 32024542 PMCID: PMC7003432 DOI: 10.1186/s12974-020-1712-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/13/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Astrocytes are the most abundant glial cells in a brain that mediate inflammatory responses and provide trophic support for neurons. We have previously disclosed that paroxetine, a common selective serotonin reuptake inhibitor, ameliorates LPS-induced microglia activation. However, it remains elusive for the role of paroxetine in astrocytic responses. METHODS Isolated primary astrocytes were pretreated with paroxetine and stimulated with different stimuli, lipopolysaccharide (LPS) or microglia conditioned medium pre-activated with LPS (M/Lps). Inflammatory and neurotrophic responses, underlying mechanisms and the impact on neuronal survival were assessed. RESULTS Paroxetine had no impact on LPS-stimulated iNOS, TNF-α, and IL-1β expression, but inhibited M/Lps-induced TNF-α and IL-1β expression in primary astrocytes. Paroxetine suppressed M/Lps- but not LPS-induced activation of NF-κB and had no impact on the activation of MAPKs and STAT3. Incubation with the resulted astrocyte conditioned media caused no change in the viability of SH-SY5Y cells. BDNF and MANF mRNA expressions were upregulated by M/Lps and paroxetine, respectively. However, M/Lps- or LPS-induced extracellular releases of NO, TNF-α, and/or BDNF in astrocytes were in minor amount compared to those by microglia. CONCLUSIONS Paroxetine ameliorates the reactive microglia-mediated inflammatory responses in astrocytes partially via inhibition of the NF-κB pathway but has no impact on LPS-stimulated astrocyte activation. While the effects of paroxetine on secondary astrocytic responses are not robust compared to its effect on the innate immune responses of microglia, the results together may implicate a therapeutic potential of paroxetine against neuroinflammation-associated neurological disorders such as Parkinson's disease.
Collapse
Affiliation(s)
- Xiong Zhang
- Department of Geriatrics and Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.,Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Lan-Bing Zhu
- Department of Geriatrics and Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Jia-Hui He
- Department of Geriatrics and Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Hong-Qiu Zhang
- Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Shu-Ya Ji
- Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Chao-Nan Zhang
- Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Na-Na Hou
- Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Chen-Ping Huang
- Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Jian-Hong Zhu
- Department of Geriatrics and Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China. .,Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
44
|
Wang JL, Xu CJ. Astrocytes autophagy in aging and neurodegenerative disorders. Biomed Pharmacother 2019; 122:109691. [PMID: 31786465 DOI: 10.1016/j.biopha.2019.109691] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 11/11/2019] [Accepted: 11/20/2019] [Indexed: 12/13/2022] Open
Abstract
Astrocytes can serve multiple functions in maintaining cellular homeostasis of the central nervous system (CNS), and normal functions for autophagy in astrocytes is considered to have very vital roles in the pathogenesis of aging and neurodegenerative diseases. Autophagy is a major intracellular lysosomal (or its yeast analog, vacuolar) clearance pathways involved in the degradation and recycling of long-lived proteins, oxidatively damaged proteins and dysfunctional organelles by lysosomes. Current evidence has shown that autophagy might influence inflammation, oxidative stress, aging and function of astrocytes. Although the interrelation between autophagy and inflammation, oxidative stress, aging or neurological disorders have been addressed in detail, the influence of astrocytes mediated-autophagy in aging and neurodegenerative disorders has yet to be fully reviewed. In this review, we will summarize the most up-to-date findings and highlight the role of autophagy in astrocytes and link autophagy of astrocytes to aging and neurodegenerative diseases. Due to the prominent roles of astrocytic autophagy in age-related neurodegenerative diseases, we believe that we can provide new suggestions for the treatment of these disorders.
Collapse
Affiliation(s)
- Jun-Ling Wang
- Center for Reproductive Medicine, Affiliated Hospital 1 of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, PR China.
| | - Chao-Jin Xu
- Department of Histology & Embryology, School of Basic Medical Science, Wenzhou Medical University, Cha Shan University Town, No.1 Central North Road, Wenzhou, Zhejiang, 325035, PR China.
| |
Collapse
|
45
|
Kaur N, Fang YC, Lee HY, Singh A, Nepali K, Lin MH, Yeh TK, Lai MJ, Chan L, Tu YK, Banerjee S, Hu CJ, Liou JP. Protective effects of 10,11-dihydro-5H-dibenzo[b,f]azepine hydroxamates on vascular cognitive impairment. Eur J Med Chem 2019; 187:111915. [PMID: 31838329 DOI: 10.1016/j.ejmech.2019.111915] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 11/24/2019] [Accepted: 11/25/2019] [Indexed: 02/07/2023]
Abstract
A series of 10,11-dihydro-5H-dibenzo [b,f]azepine hydroxamates (4-15) were synthesized, behaving as histone deacetylase inhibitors, and examined for their influence on vascular cognitive impairment (VCI), which correlated with dementia. The results revealed that (E)-3-(4-(((3-(3-chloro-10,11-dihydro-5H-dibenzo [b,f]azepin-5-yl)propyl)amino)methyl)phenyl)-N-hydroxy-acrylamide (13) increases cerebral blood flow (CBF), attenuates cognitive impairment, and improves hippocampal atrophy in in vivo study. It is also able to increase the level of histone acetylation (H3K14 or H4K5) in the cortex and hippocampus of chronic cerebral hypoperfusion (CCH) mice; as a result, it could be a potential HDAC inhibitor for the treatment of vascular cognitive impairment.
Collapse
Affiliation(s)
- Navdeep Kaur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Yao-Ching Fang
- Taipei Neuroscience Institute, Taipei Medical University, Taiwan
| | - Hsueh-Yun Lee
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Arshdeep Singh
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Mei-Hsiang Lin
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institute, Zhunan Town, Miaoli County, Taiwan
| | - Mei-Jung Lai
- TMU Biomedical Commercialization Center, Taipei Medical University, Taiwan
| | - Lung Chan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yong-Kwang Tu
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Taipei Neuroscience Institute, Taipei Medical University, Taiwan
| | - Suddhasatwa Banerjee
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Chaur-Jong Hu
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Taipei Neuroscience Institute, Taipei Medical University, Taiwan.
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan; TMU Biomedical Commercialization Center, Taipei Medical University, Taiwan.
| |
Collapse
|
46
|
Exposure to Antidepressant Medication and the Risk of Incident Dementia. Am J Geriatr Psychiatry 2019; 27:1177-1188. [PMID: 31235427 DOI: 10.1016/j.jagp.2019.05.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/27/2019] [Accepted: 05/27/2019] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To test competing hypotheses that monotherapeutic antidepressant exposure is associated with an increased versus a decreased risk of dementia. METHODS A prospective national matched cohort study from Israel (N = 71,515) without dementia (2002-2012) aged 60 and over were followed up for incident dementia from May 2013 to October 2017. Exposure to antidepressant monotherapy was classified with Anatomical Therapeutic Chemical Codes (N06A) from January 1, 2013 to December 31, 2016. The association between antidepressant monotherapy and the risk of incident dementia was quantified with hazard ratios (HR) and their 95% confidence intervals (CI) obtained from Cox regression models unadjusted and adjusted for 42 covariates. The robustness of the results was tested with 24 sensitivity analyses: 19 analyses restricted to subsamples with plausible differential dementia risks (e.g., anxiety and depression), and 5 analyses across and within antidepressant drug classes. RESULTS In the primary analysis, the risk of incident dementia for the group exposed to antidepressant monotherapy compared to the group unexposed to antidepressants was estimated with an unadjusted HR = 4.09 (df = 1, 95% Wald CI = 3.64, 4.60) and an adjusted HR = 3.43 (df = 1, 95% Wald CI = 3.04, 3.88). Across the 24 sensitivity analyses the estimated adjusted HR values ranged from 1.99 to 5.47. CONCLUSION In this study, monotherapeutic antidepressant exposure in old age was associated with increased incident dementia. Clinicians, caregivers, and patients may wish to consider this potentially negative consequence of antidepressant exposure and aim to balance the costs and benefits of treatment.
Collapse
|
47
|
Lipponen A, Natunen T, Hujo M, Ciszek R, Hämäläinen E, Tohka J, Hiltunen M, Paananen J, Poulsen D, Kansanen E, Ekolle Ndode-Ekane X, Levonen AL, Pitkänen A. In Vitro and In Vivo Pipeline for Validation of Disease-Modifying Effects of Systems Biology-Derived Network Treatments for Traumatic Brain Injury-Lessons Learned. Int J Mol Sci 2019; 20:ijms20215395. [PMID: 31671916 PMCID: PMC6861918 DOI: 10.3390/ijms20215395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/19/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
We developed a pipeline for the discovery of transcriptomics-derived disease-modifying therapies and used it to validate treatments in vitro and in vivo that could be repurposed for TBI treatment. Desmethylclomipramine, ionomycin, sirolimus and trimipramine, identified by in silico LINCS analysis as candidate treatments modulating the TBI-induced transcriptomics networks, were tested in neuron-BV2 microglial co-cultures, using tumour necrosis factor α as a monitoring biomarker for neuroinflammation, nitrite for nitric oxide-mediated neurotoxicity and microtubule associated protein 2-based immunostaining for neuronal survival. Based on (a) therapeutic time window in silico, (b) blood-brain barrier penetration and water solubility, (c) anti-inflammatory and neuroprotective effects in vitro (p < 0.05) and (d) target engagement of Nrf2 target genes (p < 0.05), desmethylclomipramine was validated in a lateral fluid-percussion model of TBI in rats. Despite the favourable in silico and in vitro outcomes, in vivo assessment of clomipramine, which metabolizes to desmethylclomipramine, failed to demonstrate favourable effects on motor and memory tests. In fact, clomipramine treatment worsened the composite neuroscore (p < 0.05). Weight loss (p < 0.05) and prolonged upregulation of plasma cytokines (p < 0.05) may have contributed to the worsened somatomotor outcome. Our pipeline provides a rational stepwise procedure for evaluating favourable and unfavourable effects of systems-biology discovered compounds that modulate post-TBI transcriptomics.
Collapse
Affiliation(s)
- Anssi Lipponen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland.
| | - Teemu Natunen
- Institute of Biomedicine, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland.
| | - Mika Hujo
- School of Computing, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland.
| | - Robert Ciszek
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland.
| | - Elina Hämäläinen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland.
| | - Jussi Tohka
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland.
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland.
| | - Jussi Paananen
- Institute of Biomedicine, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland.
- Bioinformatics Center, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland.
| | - David Poulsen
- Jacobs School of Medicine and Biomedical Sciences, University of Buffalo, 875 Ellicott St, 6071 CTRC, Buffalo, NY 14203, USA.
| | - Emilia Kansanen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland.
| | - Xavier Ekolle Ndode-Ekane
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland.
| | - Anna-Liisa Levonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland.
| | - Asla Pitkänen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland.
| |
Collapse
|
48
|
McMurray KMJ, Vollmer LL, Ahlbrand R, Thomas J, Winter A, Lewkowich IP, Sah R. Immunomodulatory T cell death associated gene-8 (TDAG8) receptor in depression-associated behaviors. Physiol Behav 2019; 209:112598. [PMID: 31271833 DOI: 10.1016/j.physbeh.2019.112598] [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: 03/22/2019] [Revised: 06/26/2019] [Accepted: 06/29/2019] [Indexed: 12/27/2022]
Abstract
Converging evidence supports neuroimmune factors in depression psychopathology. We previously reported reduced depression-like behavior in immunomodulatory G-protein-coupled receptor, T cell death-associated gene-8 (TDAG8) deficient mice. Here, we expand on those findings by investigating depression- and anxiety-associated behaviors, and cytokine profiles in TDAG8-deficient mice. TDAG8-deficiency reduced depression- and anxiety-associated behaviors in the forced swim test (FST), open-field test and elevated zero maze. Interestingly, cytokine expression, particularly IL-6, was attenuated within hippocampus and spleen in TDAG8-deficient mice following the FST. There were no differences in immune-cell frequencies. Collectively, these data suggest a contributory role of TDAG8 in neuroimmune regulation and depression-associated physiology.
Collapse
Affiliation(s)
- Katherine M J McMurray
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Lauren Larke Vollmer
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Rebecca Ahlbrand
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Joshua Thomas
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Andrew Winter
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Neuroscience Graduate Program, University of Cincinnati, Medical Sciences Building, Room 1058B, 231 Albert Sabin Way, Cincinnati, OH 45237, USA
| | - Ian P Lewkowich
- Division of Immunobiology, Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Renu Sah
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Neuroscience Graduate Program, University of Cincinnati, Medical Sciences Building, Room 1058B, 231 Albert Sabin Way, Cincinnati, OH 45237, USA; VA Medical Center, Cincinnati, OH 45237, USA
| |
Collapse
|
49
|
Interleukin-4 signalling pathway underlies the anxiolytic effect induced by 3-deoxyadenosine. Psychopharmacology (Berl) 2019; 236:2959-2973. [PMID: 30963194 DOI: 10.1007/s00213-019-5186-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/30/2019] [Indexed: 12/20/2022]
Abstract
RATIONALE Converging evidence suggests that neuroimmunity plays an important role in the pathophysiology of anxiety. Interleukin (IL)-4 is a key cytokine regulating neuroimmune functions in the central nervous system. More efficient anxiolytics with neuro-immune mechanisms are urgently needed. OBJECTIVE To determine whether 3'-deoxyadenosine (3'-dA) exerts an anxiolytic effect and to examine the role of IL-4 in the anxiolytic effect of 3'-dA in mice. METHODS We investigated the effects of 3'-dA on anxiety-like behaviors using elevated plus maze (EPM) or light-dark box (LDB) tests after 45 min or 5 days of treatment. Expression of IL-4, IL-10, IL-1β, TNF-α, and IL-6 in the prefrontal cortex (PFC) was detected by Western blot and/or double immunostaining. Intracerebroventricular injection of RIL-4Rα (an IL-4-specific inhibitor) and intraperitoneal injection of 3'-dA or imipramine were co-administered, followed by EPM test. RESULTS 3'-dA exhibited a stronger and faster anxiolytic effect than imipramine in behavioral tests. Furthermore, 3'-dA enhanced IL-4 expression after 45 min or 5 days, TNF-α and IL-1β expression decreased significantly after a 5-day treatment with 3'-dA, and IL-10 expression increased after a 5-day treatment with 3'-dA or imipramine in the PFC. IL-4 was expressed in neurons and in some astrocytes and microglia. IL-4 expression showed a strong positive correlation with reduced anxiety behaviors. RIL-4Rα completely blocked the anxiolytic effects induced by 3'-dA and imipramine. CONCLUSIONS This study identifies a novel and common anxiolytic IL-4 signaling pathway and provides an innovative drug with a novel neuro-immune mechanism for treating anxiety disorder.
Collapse
|
50
|
Dopaminergic impact of cART and anti-depressants on HIV neuropathogenesis in older adults. Brain Res 2019; 1723:146398. [PMID: 31442412 DOI: 10.1016/j.brainres.2019.146398] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 08/09/2019] [Accepted: 08/19/2019] [Indexed: 01/21/2023]
Abstract
The success of combination antiretroviral therapy (cART) has transformed HIV infection into a chronic condition, resulting in an increase in the number of older, cART-treated adults living with HIV. This has increased the incidence of age-related, non-AIDS comorbidities in this population. One of the most common comorbidities is depression, which is also associated with cognitive impairment and a number of neuropathologies. In older people living with HIV, treating these overlapping disorders is complex, often creating pill burden or adverse drug-drug interactions that can exacerbate these neurologic disorders. Depression, NeuroHIV and many of the neuropsychiatric therapeutics used to treat them impact the dopaminergic system, suggesting that dopaminergic dysfunction may be a common factor in the development of these disorders. Further, changes in dopamine can influence the development of inflammation and the regulation of immune function, which are also implicated in the progression of NeuroHIV and depression. Little is known about the optimal clinical management of drug-drug interactions between cART drugs and antidepressants, particularly in regard to dopamine in older people living with HIV. This review will discuss those interactions, first examining the etiology of NeuroHIV and depression in older adults, then discussing the interrelated effects of dopamine and inflammation on these disorders, and finally reviewing the activity and interactions of cART drugs and antidepressants on each of these factors. Developing better strategies to manage these comorbidities is critical to the health of the aging, HIV-infected population, as the older population may be particularly vulnerable to drug-drug interactions affecting dopamine.
Collapse
|