1
|
Wu T, Xiong Y, Wang L, Wu J, Yin Y, Wang M. Accelerated aging mediates the association between rheumatoid arthritis and depression severity. J Affect Disord 2025; 379:861-869. [PMID: 40032139 DOI: 10.1016/j.jad.2025.02.096] [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: 08/16/2024] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND This study aimed to investigate the associations among phenotypic age acceleration (PAA), the prevalence of rheumatoid arthritis (RA), and depression severity and to examine the potential mediating role of PAA in the RA-depression relationship. METHODS A retrospective cohort analysis was conducted using data from adult participants in the National Health and Nutrition Examination Survey (2005-2010). Participants with RA were identified on the basis of self-report of RA in personal interviews. Depression was assessed via the Patient Health Questionnaire-9 (PHQ-9). PAA was calculated with 9 clinical biomarkers and chronological age, with a relatively high RAA value indicating the acceleration of aging. Survey-weighted logistic regression models were used to explore the relationship between the prevalence of RA and PAA and the correlation between PAA and depression. Mediation analysis was employed to quantify the indirect effect of RA on depression through PAA. RESULTS A total of 9834 adults aged over 20 years were included. RA prevalence was positively associated with greater PAA (β = 1.60, 95 % confidence interval (CI): 0.86-2.35, P = 0.0002) and depression severity (β = 1.57, 95 % CI: 1.09-2.05, P < 0.0001). Additionally, PAA was correlated with depression severity (β = 0.04, 95 % CI: 0.02-0.06; P = 0.0001). Mediation analysis revealed that PAA significantly mediated the association between RA and depressive severity, explaining 2.26-5.57 % (all p < 0.001) of the total effect. CONCLUSIONS Individuals with RA exhibited accelerated biological aging, as represented by increased PAA, which partially mediated the relationship between RA and depression severity in this population.
Collapse
Affiliation(s)
- Tingting Wu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Soochow University, No.188 Shizi St, Suzhou 215006, China
| | - Yue Xiong
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Soochow University, No.188 Shizi St, Suzhou 215006, China
| | - Lei Wang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Soochow University, No.188 Shizi St, Suzhou 215006, China
| | - Jian Wu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Soochow University, No.188 Shizi St, Suzhou 215006, China
| | - Yufeng Yin
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Soochow University, No.188 Shizi St, Suzhou 215006, China.
| | - Mingjun Wang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Soochow University, No.188 Shizi St, Suzhou 215006, China.
| |
Collapse
|
2
|
Barichello T, Kluwe-Schiavon B, Borba LA, Pedro LC, Niero FS, Dos Santos LN, Leonardo LM, Ignácio ZM, Morales R, Ceretta LB, Reus GZ. Alterations in Gut Microbiome Composition and Increased Inflammatory Markers in Post-COVID-19 Individuals. Mol Neurobiol 2025; 62:8038-8047. [PMID: 39966327 PMCID: PMC12078008 DOI: 10.1007/s12035-025-04769-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
Dysfunctions in the immune system and alterations in the microbiome composition following SARS-CoV-2 infection contribute to persistent neurological issues observed in long COVID-19 survivors. We hypothesize that alterations in the gut microbiome composition and peripheral inflammatory profile following COVID-19 may play pivotal roles in behavior changes among individuals experiencing long-term illness. This cross-sectional study included a sample of post-COVID-19 and non-COVID-19 subjects. We assessed the presence of psychiatric conditions utilizing standardized diagnostic criteria, Hamilton Rating Scale for Anxiety (HAM-A), Hamilton Rating Scale for Depression (HAM-D), Biological Rhythms in Neuropsychiatry Assessment Interview (BRIAN), and Functional Assessment Short Test (FAST). Plasma samples were analyzed to examine lipid and inflammatory profiles. Fecal samples were evaluated by 16S rRNA sequencing to identify the gut microbiome composition. Noteworthy findings include a significant increase in the myeloid progenitor inhibitory factor 1 (MPIF-1), interleukin (IL)-17, and triglyceride among post-COVID-19 individuals. While α-diversity in the gut microbiome composition showed no significant differences, β-diversity demonstrated a notable distinction between the healthy control and post-COVID-19 groups. Post-COVID-19 individuals exhibited a decreased abundance of phylum, class, and order of Verrucomicrobia, family, and genus of Akkermansia, a short-chain fatty acid producer and microbial group significantly associated with intestinal barrier homeostasis and the amelioration of metabolic diseases. No difference was found between the behavioral and clinical data. In post-COVID-19 individuals, there were elevated IL-17 and MPIF-1 levels, compared to non-COVID-19 individuals. Additionally, there were notable alterations in gut microbiome composition, as evidenced by changes in β-diversity and a decrease of Verrucomicrobia, family, and Akkermansia genus abundance.
Collapse
Affiliation(s)
- Tatiana Barichello
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, Mcgovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Neuroscience Graduate Program, the University of Texas MD Anderson Cancer Center Uthealth Graduate School of Biomedical Sciences, Houston, Texas, USA
- Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Bruno Kluwe-Schiavon
- Department of Psychiatry and Behavioral Sciences, Mcgovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Laura A Borba
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Lucas C Pedro
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Flávia S Niero
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Laísa N Dos Santos
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Letícia M Leonardo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Zuleide M Ignácio
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of the Southern Frontier, Chapecó, SC, Brazil
| | - Rodrigo Morales
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston (Uthealth), Houston, Texas, USA
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| | - Luciane B Ceretta
- Graduate Program in Public Health, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Gislaine Z Reus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
3
|
Bi H, Jin J, Sun M, Chen M, Li X, Wang Y. Epigenetic changes caused by early life stress in the pathogenesis of depression. Eur J Pharmacol 2025; 999:177671. [PMID: 40288560 DOI: 10.1016/j.ejphar.2025.177671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 04/16/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
Major depressive disorder (MDD) is a severe psychiatric disorder with a complex and poorly understood pathogenesis. Epigenetics, a rapidly advancing field of biology, has been implicated in various psychiatric conditions, including schizophrenia, anxiety, substance addiction, and autism. Furthermore, substantial research indicates that epigenetic modifications play a crucial role in the etiology of depression. Early life stress (ELS) refers to adverse experiences occurring during prenatal development (e.g., maternal physical and mental health complications during pregnancy) and/or postnatal life (e.g., abuse, neglect, poverty, parental loss, family conflict, violence, and malnutrition). These early-life adversities can lead to epigenetic modifications, which, in turn, influence key biological processes and contribute to the pathogenesis of MDD. This review provides an overview of the regulatory mechanisms and functions of various epigenetic modifications, including non-coding RNAs, DNA methylation, and histone modifications. We then examine ELS-induced epigenetic alterations and their biological consequences, such as dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, neurogenesis, and neuroplasticity. Finally, we explore their potential implications for both the pathogenesis and treatment of MDD. We hypothesize that ELS-induced epigenetic changes may serve as biomarkers for MDD diagnosis and offer novel therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Hongsheng Bi
- Department of Psychiatry, The First Hospital of China Medical University, China; The Third Hospital of Daqing, Psychiatric Ward No. 2, China
| | - Jingyan Jin
- Department of Psychiatry, The First Hospital of China Medical University, China
| | - Mingyuan Sun
- Department of Psychiatry, The First Hospital of China Medical University, China
| | - Mingrui Chen
- Department of Psychiatry, The First Hospital of China Medical University, China
| | - Xiaobai Li
- Department of Psychiatry, The First Hospital of China Medical University, China.
| | - Yan Wang
- Center for Psychological Development, China Medical University, China.
| |
Collapse
|
4
|
Tang L, Wu L, Dai M, Liu N, Liu L. Integrative analysis of signaling and metabolic pathways, immune infiltration patterns, and machine learning-based diagnostic model construction in major depressive disorder. Sci Rep 2025; 15:13519. [PMID: 40253457 PMCID: PMC12009401 DOI: 10.1038/s41598-025-97623-x] [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: 10/10/2024] [Accepted: 04/07/2025] [Indexed: 04/21/2025] Open
Abstract
Major depressive disorder (MDD) is a multifactorial disorder involving genetic and environmental factors, with unclear pathogenesis. This study aims to explore the pathogenic pathway of MDD and its relationship with immune responses and to discover its potential targets by bioinformatics methods. We first applied gene set variation analysis (GSVA) and seven different immune infiltration algorithms to the GSE98793 dataset to determine the differences in signaling pathways, metabolic pathways, and immune cell infiltration between MDD patients and healthy controls. Differentially expressed genes between MDD patients and controls were obtained from five datasets (GSE98793, GSE32280, GSE38206, GSE39653, and GSE52790), and 113 machine learning methods were employed to construct MDD diagnostic models. Based on the constructed MDD diagnostic models, MDD patients were divided into high-risk and low-risk groups. GSVA and immune microenvironment analyses were conducted to investigate the differences between the two groups. Furthermore, potential drugs and therapeutic targets for the high-risk MDD group were explored to provide new insights and directions for the precise treatment of MDD. GSVA and immune infiltration results indicate that patients with MDD exhibit differences from normal individuals in various aspects, including biological processes, signaling pathways, metabolic processes, and immune cells. To investigate the functions and biological significance of differentially expressed genes in MDD patients, we performed GO and KEGG enrichment analyses on the differentially expressed genes from five databases (GSE98793, GSE32280, GSE38206, GSE39653, and GSE52790). By comparing the enrichment results across the five datasets, we found that the cell-killing signaling pathway was consistently present in the enriched signaling pathways of all datasets, suggesting that this pathway may play a crucial role in the pathogenesis of MDD. The random forest algorithm (AUC = 0.788) was selected as the optimal algorithm from 113 machine learning algorithms, leading to the development of a robust and predictive MDD algorithm, highlighting the important role of NPL in MDD. By dividing MDD into high and low-risk subgroups based on diagnostic model scores, enrichment pathways, and immunological results further demonstrated that high-risk MDD is associated with increased levels of reactive oxygen species, inflammation, and numbers of T cells and B cells. Through GSEA scoring, five upregulated pathways in the high-risk MDD group were identified, and multiple potential drugs such as Mibefradil, LY364947, ZLN005, STA- 5326, and vemurafenib were screened. Patients with MDD show differences in signaling pathways, metabolic pathways, and immune mechanisms. By constructing an MDD diagnostic model, we predicted the key genes of MDD and the characteristic pathways associated with a higher risk of MDD. This provides new insights for risk stratification identification and offers new perspectives for the clinical application of precision immunotherapy and drug development.
Collapse
Affiliation(s)
- Lei Tang
- Mental Health Center, Affiliated Hospital of North Sichuan Medical College, 1 South Maoyuan Road, 637000, Nanchong, China
- School of Psychiatry, North Sichuan Medical College, Nanchong, China
- Department of Psychiatry, Sleep Medicine Centre, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Liling Wu
- Department of Pharmacy, The Second Clinical School of North Sichuan Medical College, Nanchong Hospital of Beijing Anzhen Hospital CMU (Nanchong Central Hospital), Nanchong, China
| | - Mengqin Dai
- Mental Health Center, Affiliated Hospital of North Sichuan Medical College, 1 South Maoyuan Road, 637000, Nanchong, China
- School of Psychiatry, North Sichuan Medical College, Nanchong, China
| | - Nian Liu
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China.
| | - Lu Liu
- Mental Health Center, Affiliated Hospital of North Sichuan Medical College, 1 South Maoyuan Road, 637000, Nanchong, China.
- School of Psychiatry, North Sichuan Medical College, Nanchong, China.
| |
Collapse
|
5
|
Cao Y, Fan X, Zang T, Li Y, Tu Y, Wei Y, Bai J, Liu Y. Gut microbiota causes depressive phenotype by modulating glycerophospholipid and sphingolipid metabolism via the gut-brain axis. Psychiatry Res 2025; 346:116392. [PMID: 39933221 DOI: 10.1016/j.psychres.2025.116392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/26/2025] [Accepted: 02/06/2025] [Indexed: 02/13/2025]
Abstract
Emerging evidence suggests that changes in the gut microbiota (GM) are related to prenatal depression onset, but the underlying molecular mechanisms remain obscure. This study was conducted to explore how disordered GM is involved in the onset of prenatal depression through the microbiome-gut-brain (MGB) axis. We transplanted fecal microbiota from women with and without prenatal depression into germ-free mice. Fecal metagenomic sequencing and LC-MS untargeted metabolomics analysis were performed to identify the GM composition, function, and metabolites in mice. Lipid metabolomics analysis was then used to characterize the lipid metabolism of brain tissue in mice. We found that mice transplanted with fecal microbiota from women with prenatal depression exhibited depressive-like behaviors as well as characteristic disorders of the phylum Firmicutes. Weighted Gene Correlation Network Analysis identified three microbial and one metabolic module in the gut, alongside two lipid metabolic modules in the brain, as significantly related to all depressive-like behaviors. These modules were enriched for glycerophospholipid and sphingolipid metabolism. In addition, the GM of mice with depressive-like behaviors were enriched and deficient in relevant functions and enzymes in the glycerophospholipid (mainly phosphatidylethanolamine) and sphingolipid (mainly hexosyl-ceramide) metabolic pathways, respectively. Consistently, glycerophospholipid and sphingolipid metabolites in the brains of depressive-like mice were up- and down-regulated. Increased phosphatidylethanolamine and decreased hexosyl-ceramide were significantly related to differential genera in the gut. Collectively, our findings provide a novel microbial and metabolic framework for understanding the role of the MGB axis in prenatal depression, indicating that the GM may be involved in the onset of depressive phenotypes by modulating central glycerophospholipid and sphingolipid metabolic homeostasis.
Collapse
Affiliation(s)
- Yanan Cao
- Renmin Hospital of Wuhan University, Wuhan, 430060, PR China
| | - Xiaoxiao Fan
- Center for Women's and Children's Health, Wuhan University School of Nursing, Wuhan University, Wuhan, 430071, PR China
| | - Tianzi Zang
- Center for Women's and Children's Health, Wuhan University School of Nursing, Wuhan University, Wuhan, 430071, PR China
| | - Yanting Li
- Center for Women's and Children's Health, Wuhan University School of Nursing, Wuhan University, Wuhan, 430071, PR China
| | - Yiming Tu
- Center for Women's and Children's Health, Wuhan University School of Nursing, Wuhan University, Wuhan, 430071, PR China
| | - Yi Wei
- Center for Women's and Children's Health, Wuhan University School of Nursing, Wuhan University, Wuhan, 430071, PR China
| | - Jinbing Bai
- Emory University Nell Hodgson Woodruff School of Nursing, 1520 Clifton Road, Atlanta, GA, 30322, USA
| | - Yanqun Liu
- Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Center for Women's and Children's Health, Wuhan University School of Nursing, Wuhan University, Wuhan, 430071, PR China.
| |
Collapse
|
6
|
Wang H, Peng J, Zhu H, Sun M, Fang Y, Chen Z, Hu W, Tong L, Ren J, Huang C, Lu X. Microglia stimulation produces antidepressant-like effects in a mouse depression model induced by adolescent chronic unpredictable stress. Physiol Behav 2025; 291:114782. [PMID: 39672484 DOI: 10.1016/j.physbeh.2024.114782] [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: 06/02/2024] [Revised: 11/25/2024] [Accepted: 12/10/2024] [Indexed: 12/15/2024]
Abstract
Depression triggered by harmful stress in adolescents is a common phenomenon that can lead to serious social problems. Its treatment is still frustrating in the clinic. We reported the phenomenon that a 12-day chronic unpredictable stress (CUS), starting on postnatal day 28, led to a significant decrease in the number of microglia in the dentate gyrus of the hippocampus in adult mice. Reversing this decline with a single injection of low-dose lipopolysaccharide (LPS), a classical immunostimulant, could rapidly reverse the depression-like behavior induced by 12 days of CUS stimulation during adolescence. In the dose-dependent experiment, a single injection of LPS at doses of 75 and 100 μg/kg, but not at doses of 25 and 50 μg/kg, produced an antidepressant effect in mice exposed to 12-day CUS during adolescence. The time-dependent experiment showed that the antidepressant effect of the single injection of LPS (100 μg/kg) occurred at time points 5 and 8 h, but not 3 h after LPS injection. The antidepressant effect of the single LPS injection (100 μg/kg) lasted for at least 7 days, and 14 days after the single LPS injection, a repeated injection could still induce the stressed mice to develop an antidepressant phenotype. Furthermore, inhibition of microglia by minocycline or depletion of microglia by PLX3397 was found to prevent the antidepressant effect of the single LPS injection. These results suggest that reversing the decline of microglia in the dentate gyrus may be a potential strategy for the treatment of depression induced by harmful stress in adolescents.
Collapse
Affiliation(s)
- Hanxiao Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China
| | - Jie Peng
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China
| | - Haojie Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China
| | - Micona Sun
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China
| | - Yunli Fang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China
| | - Zhuo Chen
- Invasive Technology Department, Nantong First People's Hospital, The Second Affiliated Hospital of Nantong University, #6 North Road Hai'er Xiang, Nantong 226001, Jiangsu Province, China
| | - Wenfeng Hu
- Department of Pharmacy, Affiliated Maternal and Child Health Hospital of Nantong University, #399 Shijidadao, Nantong 226007, China
| | - Lijuan Tong
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China
| | - Jie Ren
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China.
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China.
| |
Collapse
|
7
|
Rangel MVDS, Lopes KG, Qin X, Borges JP. Exercise-induced adaptations in the kynurenine pathway: implications for health and disease management. Front Sports Act Living 2025; 7:1535152. [PMID: 40115437 PMCID: PMC11922725 DOI: 10.3389/fspor.2025.1535152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/25/2025] [Indexed: 03/23/2025] Open
Abstract
Background Tryptophan (TRP) metabolism through the kynurenine (KYN) pathway is influenced by inflammatory mediators, generating metabolites that regulate immune and inflammatory responses. Exercise has been proposed as a modulator of this pathway, but its role in health benefits and chronic disease management remains unclear. Objective This systematic review examines exercise-induced adaptations in the KYN pathway and their potential implications for health and disease management. Additionally, we identify key methodological considerations for future research. Methods A structured search of PubMed/Medline, Web of Science, and Scopus was conducted up to October 2024 to identify clinical trials investigating the effects of exercise training on the KYN pathway. Results Of 2,795 articles initially found, 13 clinical trials involving 592 participants met the inclusion criteria. Most studies reported exercise-induced adaptations in the KYN pathway, particularly in cancer survivors. These adaptations appeared to be influenced by exercise intensity and duration. However, several methodological limitations were noted, and no trials included patients with metabolic or cardiovascular diseases. Conclusions Here, we show that exercise training modulates the KYN pathway in both healthy and diseased populations, highlighting its potential for disease prevention and management. However, further randomized-controlled trials are needed to clarify its mechanisms and clinical applications, particularly in metabolic and cardiovascular diseases. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO/view/CRD42022351481, PROSPERO (CRD42022351481).
Collapse
Affiliation(s)
- Marcus Vinicius Dos Santos Rangel
- Laboratory of Physical Activity and Health Promotion, Institute of Physical Education and Sports, University of Rio de Janeiro State, Rio de Janeiro, RJ, Brazil
| | - Karynne Grutter Lopes
- Postgraduate Program in Clinical and Experimental Physiopathology, Faculty of Medical Sciences, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Xuebin Qin
- Department of Microbiology and Immunology, Tulane National Primate Research Center and Tulane University School of Medicine, Covington, LA, United States
| | - Juliana Pereira Borges
- Laboratory of Physical Activity and Health Promotion, Institute of Physical Education and Sports, University of Rio de Janeiro State, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
8
|
Bostan ZZ, Şare Bulut M, Gezmen Karadağ M. Can Vıtamın D Reduce the Need for SSRI by Modulatıng Serotonın Synthesıs?: A Revıew of Recent Lıterature. Curr Nutr Rep 2025; 14:39. [PMID: 40025236 PMCID: PMC11872774 DOI: 10.1007/s13668-025-00630-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2025] [Indexed: 03/04/2025]
Abstract
PURPOSE OF REVIEW This study aims to analyze the effects of vitamin D on serotonin synthesis, release, and function in the brain, and to explore its relationship with various psychiatric disorders. RECENT FINDINGS The hormone calcitriol plays a vital role in regulating a multitude of biological processes within the human body. Its deficiency can lead to significant adverse effects on overall health and well-being, including physical and psychological consequences. Evidence indicates that vitamin D may exert influences through receptors in the brain, modulating serotonin production and influencing emotional and cognitive processes. Recent studies propose that elevated serum vitamin D may enhance mood and alleviate depressive states. The impact of vitamin D on cognitive function and behavior remains an area of ongoing investigation. This literature review analyses the available evidence on how vitamin D intake impacts the severity of illness and medication requirements in diverse psychiatric disorders. A review of the literature suggests that there may be a correlation between vitamin D and serotonin, which could potentially contribute to more favorable outcomes in the context of illness. Vitamin D may increase the amount of serotonin in the synaptic gap, which is the intended use of selective serotonin reuptake inhibitors (SSRIs), with its effect on the increase in serotonin release. According to reports, vitamin D supplementation in conjunction with SSRI group medication provides an additive effect for the management of psychiatric disorders.
Collapse
Affiliation(s)
- Zekiye Zeynep Bostan
- Department of Nutrition and Dietetics, Faculty of Health Sciences, İstanbul Gedik University, Istanbul, Türkiye
| | - Melike Şare Bulut
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Biruni University, Istanbul, Türkiye.
| | - Makbule Gezmen Karadağ
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara, Türkiye
| |
Collapse
|
9
|
Laurent N, Bellamy EL, Hristova D, Houston A. Ketogenic metabolic therapy in the remission of chronic major depressive disorder: a retrospective case study. Front Nutr 2025; 12:1549782. [PMID: 40083888 PMCID: PMC11903285 DOI: 10.3389/fnut.2025.1549782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 02/13/2025] [Indexed: 03/16/2025] Open
Abstract
Background There is limited evidence describing the use of ketogenic metabolic therapy (KMT), also known as a ketogenic diet (KD), to achieve full remission of treatment-resistant major depressive disorder (MDD) in real-world clinical settings. This case study examines a 47-year-old woman with lifelong treatment-resistant MDD who achieved complete remission of depressive symptoms and improved functioning through a ketogenic diet. Methods The patient engaged in KMT with a 1.5:1 macronutrient ratio under the supervision of a treatment team consisting of a medical professional, psychotherapist, and ketogenic-informed nutrition professional through an online program that provided both individual and group support. Interventions included dietary modifications, micronutrient supplementation, and participation in a group coaching program. Outcomes were assessed using validated tools for symptom severity, including PHQ-9 for depression and GAD-7 for anxiety, at baseline, 2 months, and 4 months post-intervention. Qualitative data on patient experiences and functional improvements were also collected. Results The patient achieved remission of MDD within 8 weeks of initiating KMT, with PHQ-9 scores decreasing from 25 (severe depression) at baseline to 0 at 2- and 4-month assessments. GAD-7 scores decreased from 3 (minimal anxiety) to 0 over the same period. Qualitative findings revealed significant improvements in emotional regulation, energy levels, and cognitive function. Conclusion This case study demonstrates the potential of KMT as a non-pharmacological intervention for achieving full remission in treatment-resistant MDD. These findings suggest further research to evaluate feasibility, efficacy, and broader applicability in diverse clinical settings.
Collapse
Affiliation(s)
| | - Erin L. Bellamy
- School of Psychology, University of East London, London, United Kingdom
| | | | - Ally Houston
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
10
|
Torres G, Subbaiah RT, Sood RA, Leheste JR. From air to mind: unraveling the impact of indoor pollutants on psychiatric disorders. Front Psychiatry 2025; 15:1511475. [PMID: 39850067 PMCID: PMC11754266 DOI: 10.3389/fpsyt.2024.1511475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/20/2024] [Indexed: 01/25/2025] Open
Abstract
Epidemiological evidence from the past 20 years indicates that environmental chemicals brought into the air by the vaporization of volatile organic compounds and other anthropogenic pollutants might be involved, at least in part, in the development or progression of psychiatric disorders. This evidence comes primarily from occupational work studies in humans, with indoor occupations being the most important sources of airborne pollutants affecting neural circuits implicated in mood disorders (e.g., major depressive disorder and bipolar disorder). The current mini review brings together recent findings of indoor airborne pollution from different fields of research, including genetics, neuropathology, and neuroimaging, for gauging underlying physiological mechanisms leading to emotional disturbances that impact nearly all aspects of human behavior. A better understanding of how indoor airborne pollutants affect brain neurons to augment clinical symptoms associated with psychiatric disorders will undoubtedly be useful in the subsequent treatment of patients with major depressive and/or bipolar disorders. This article is part of the themed issue, "Understanding the Link Between Environmental Pollutants, Brain & Behavior."
Collapse
Affiliation(s)
- German Torres
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine (NYITCOM), Old Westbury, NY, United States
| | - Ryia T. Subbaiah
- Department of Arts and Sciences, Georgetown University, Washington, DC, United States
| | - Riya A. Sood
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine (NYITCOM), Old Westbury, NY, United States
| | - Joerg R. Leheste
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine (NYITCOM), Old Westbury, NY, United States
| |
Collapse
|
11
|
Mingardi J, Meanti R, Paoli C, Cifani C, Torsello A, Popoli M, Musazzi L. Ghrelin, Neuroinflammation, Oxidative Stress, and Mood Disorders: What Are the Connections? Curr Neuropharmacol 2025; 23:172-186. [PMID: 39041263 PMCID: PMC11793048 DOI: 10.2174/1570159x22999240722095039] [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: 12/15/2023] [Revised: 03/08/2024] [Accepted: 03/19/2024] [Indexed: 07/24/2024] Open
Abstract
Ghrelin is a gut peptide hormone associated with feeding behavior and energy homeostasis. Acylated ghrelin binds to the growth hormone secretagogue receptor 1a subtype (GHS-R1a) in the hippocampus, leading to GH release from the anterior pituitary. However, in recent years, ghrelin and its receptor have also been implicated in other processes, including the regulation of cardiomyocyte function, muscle trophism, and bone metabolism. Moreover, GHS-R1a is distributed throughout the brain and is expressed in brain areas that regulate the stress response and emotional behavior. Consistently, a growing body of evidence supports the role of ghrelin in regulating stress response and mood. Stress has consistently been shown to increase ghrelin levels, and despite some inconsistencies, both human and rodent studies suggested antidepressant effects of ghrelin. Nevertheless, the precise mechanism by which ghrelin influences stress response and mood remains largely unknown. Intriguingly, ghrelin and GHS-R1a were consistently reported to exert anti-inflammatory, antioxidant, and neurotrophic effects both in vivo and in vitro, although this has never been directly assessed in relation to psychopathology. In the present review we will discuss available literature linking ghrelin with the stress response and depressive-like behavior in animal models as well as evidence describing the interplay between ghrelin and neuroinflammation/oxidative stress. Although further studies are required to understand the mechanisms involved in the action of ghrelin on mood, we hypothesize that the antiinflammatory and anti-oxidative properties of ghrelin may give a key contribution.
Collapse
Affiliation(s)
- Jessica Mingardi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Ramona Meanti
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Caterina Paoli
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Carlo Cifani
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Antonio Torsello
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Maurizio Popoli
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milano, Italy
| | - Laura Musazzi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
12
|
Gao Z, Peng J, Zhang Y, Chen Z, Song R, Song Z, Feng Q, Sun M, Zhu H, Lu X, Yang R, Huang C. Hippocampal SENP3 mediates chronic stress-induced depression-like behaviors by impairing the CREB-BDNF signaling. Neuropharmacology 2025; 262:110203. [PMID: 39486575 DOI: 10.1016/j.neuropharm.2024.110203] [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: 08/18/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Impaired signaling between cyclic adenosine monophosphate response element binding protein (CREB) and brain-derived neurotrophic factor (BDNF) in the hippocampus is generally considered to be the cause of depression. The mechanisms underlying the impairment of CREB-BDNF signaling under stress conditions are largely unclear. Small ubiquitin-like modifier (SUMO) specific peptidase 3 (SENP3) is a molecule that can regulate SUMOylation of target proteins related to synaptic plasticity. Its dynamic changes have been reported to be associated with neuronal damage in various models of central nervous disorders such as cerebral ischemia and traumatic brain injury. However, its role in depression is completely unknown. This problem was addressed in the present study. Our results showed that chronic unpredictable stress (CUS) triggered a specific increase in SENP3 expression in the hippocampus of non-stressed mice. Overexpression of SENP3 in the hippocampus of non-stressed mice elicited depression-like behaviors in the tail suspension test, forced swimming test, and sucrose preference test, accompanied by impairment of the CREB-BDNF signaling cascade in the hippocampus. Conversely, genetic silencing of SENP3 in the hippocampus suppressed the development of depression-like behaviors. Furthermore, infusion of SENP3-shRNA into the hippocampus failed to suppress CUS-induced depression-like behaviors when mice received genetic silencing CREB or BDNF in the hippocampus or inhibition of the BDNF receptor by K252a. Taken together, these results suggest that abnormally elevated SENP3 in the hippocampus leads to the development of depression-like behavior by impairing the CREB-BDNF signaling cascade. SENP3 in the hippocampus could be a promising target for the development of new antidepressants.
Collapse
Affiliation(s)
- Zhiwei Gao
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Jiangsu Province, #20 Xisi Road, Nantong 226001, Jiangsu Province, China
| | - Jie Peng
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China
| | - Yi Zhang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China
| | - Zhuo Chen
- Invasive Technology Department, Nantong First People's Hospital, The Second Affiliated Hospital of Nantong University, #6 North Road Hai'er Xiang, Nantong 226001, Jiangsu Province, China
| | - Rongrong Song
- Department of Emergency and Critical Care Medicine, Tongzhou People's Hospital, 999 Jianshe Road, Nantong 226300, Jiangsu Province, China
| | - Ze Song
- Department of Neurosurgery, Tongzhou People's Hospital, 999 Jianshe Road, Nantong 226300, Jiangsu Province, China
| | - Qijie Feng
- Department of Orthopedics, Tongzhou People's Hospital, 999 Jianshe Road, Nantong 226300, Jiangsu Province, China
| | - Micona Sun
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China
| | - Haojie Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China
| | - Rongrong Yang
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Jiangsu Province, #20 Xisi Road, Nantong 226001, Jiangsu Province, China.
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China.
| |
Collapse
|
13
|
Iriarte-Yoller N, Etxaniz-Oses J, Pavón-Navajas C, Tous-Espelosin M, Sánchez-Gómez PM, Maldonado-Martín S, Yoller-Elburgo AB, Elizagarate-Zabala E. Treatment with combined exercise in patients with resistant major depression (TRACE-RMD): study protocol for a randomised controlled trial. Trials 2024; 25:827. [PMID: 39696604 PMCID: PMC11653556 DOI: 10.1186/s13063-024-08685-7] [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: 08/24/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Around 40% of people with major depressive disorder (MDD) experience moderate remission, with the remainder meeting the criteria for resistant major depression (RMD). It has been shown that exercise has a low-to-moderate effect on MDD, but there is a lack of evidence on exercise interventions in RMD patients. The primary purpose of the proposed study will be to investigate the effect of a 12-week supervised combined exercise program on depressive symptoms in people with RMD compared to a treatment-as-usual (TAU) group. METHOD This randomised, single-blind, controlled experimental trial will include 70 adults (≥ 18 years old) with RMD. Participants randomised to an exercise intervention, or a TAU group will be assessed at baseline and after a three-month intervention period. The primary variable will be participants' depressive symptoms measured with the Montgomery-Asberg Depression Rating Scale. Secondary outcome variables will include cardiorespiratory fitness (peak oxygen uptake through peak cardiopulmonary exercise test), body composition (bioimpedance and anthropometric variables), physical activity level (the International Physical Activity Questionnaire), health-related quality of life (the Short Form-36 Health Survey), functional outcome (the Sheehan Disability Scale and Quality of Life in Depression Scale), overall disease severity (the Clinical Global Impression Scale-Severity of Illness), and biochemical variables (a fasting blood sample). DISCUSSION This study will try to answer whether a supervised co-adjuvant combined (aerobic and resistance training) exercise program will help the prognosis of this population with RMD. TRIAL REGISTRATION ClinicalTrials.gov NCT05136027. Last public release on 12/13/2023.
Collapse
Affiliation(s)
- Nagore Iriarte-Yoller
- Osakidetza Basque Health Service, Araba Mental Health Network, Psychiatric Hospital of Alava, Vitoria-Gasteiz, Spain
- Bioaraba, New Therapies in Mental Health Group, Vitoria-Gasteiz, Spain
- Department of Medicine, Faculty of Health Sciences, University of Deusto, Bilbao, Spain
| | - José Etxaniz-Oses
- Department of Physical Education and Sport, Faculty of Education and Sport-Physical Activity and Sport Sciences Section, GIzartea, Kirola eta Ariketa Fisikoa Ikerkuntza Taldea (GIKAFIT), Society, Sports, and Exercise Research Group, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Araba/Álava, Basque Country, Spain
- Bioaraba Health Research Institute, Physical Activity, Exercise, and Health group, Vitoria-Gasteiz, Basque Country, Spain
| | - Cristobal Pavón-Navajas
- Osakidetza Basque Health Service, Araba Mental Health Network, Psychiatric Hospital of Alava, Vitoria-Gasteiz, Spain
- Bioaraba, New Therapies in Mental Health Group, Vitoria-Gasteiz, Spain
- Department of Medicine, Faculty of Health Sciences, University of Deusto, Bilbao, Spain
| | - Mikel Tous-Espelosin
- Department of Physical Education and Sport, Faculty of Education and Sport-Physical Activity and Sport Sciences Section, GIzartea, Kirola eta Ariketa Fisikoa Ikerkuntza Taldea (GIKAFIT), Society, Sports, and Exercise Research Group, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Araba/Álava, Basque Country, Spain
- Bioaraba Health Research Institute, Physical Activity, Exercise, and Health group, Vitoria-Gasteiz, Basque Country, Spain
| | - Pedro M Sánchez-Gómez
- Bioaraba, New Therapies in Mental Health Group, Vitoria-Gasteiz, Spain
- Department of Medicine, Faculty of Health Sciences, University of Deusto, Bilbao, Spain
- Osakidetza Basque Health Service, Bizkaia Mental Health Network, Hospital of Zamudio, Bilbao, Spain
| | - Sara Maldonado-Martín
- Department of Physical Education and Sport, Faculty of Education and Sport-Physical Activity and Sport Sciences Section, GIzartea, Kirola eta Ariketa Fisikoa Ikerkuntza Taldea (GIKAFIT), Society, Sports, and Exercise Research Group, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Araba/Álava, Basque Country, Spain.
- Bioaraba Health Research Institute, Physical Activity, Exercise, and Health group, Vitoria-Gasteiz, Basque Country, Spain.
| | - Ana B Yoller-Elburgo
- Osakidetza Basque Health Service, Araba Mental Health Network, Psychiatric Hospital of Alava, Vitoria-Gasteiz, Spain
| | - Edorta Elizagarate-Zabala
- Osakidetza Basque Health Service, Araba Mental Health Network, Psychiatric Hospital of Alava, Vitoria-Gasteiz, Spain
- Bioaraba, New Therapies in Mental Health Group, Vitoria-Gasteiz, Spain
- Department of Medicine, Faculty of Health Sciences, University of Deusto, Bilbao, Spain
| |
Collapse
|
14
|
Chen X, Mo X, Zhang Y, He D, Xiao R, Cheng Q, Wang H, Liu L, Li WW, Xie P. A comprehensive analysis of the differential expression in the hippocampus of depression induced by gut microbiota compared to traditional stress. Gene 2024; 927:148633. [PMID: 38838871 DOI: 10.1016/j.gene.2024.148633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/22/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024]
Abstract
Depression, which is a disease of heterogeneous etiology, is characterized by high disability and mortality rates. Gut microbiota are associated with the development of depression. To further explore any differences in the mechanisms of depression induced by gut microbiota and traditional stresses, as well as facilitate the development of microbiota-based interventions, a fecal microbiota transplantation (FMT) depression model was made. This was achieved by transplanting feces from major depressive disorder (MDD) patients into germ-free mice. Second, the mechanisms of the depression induced by gut microbiota were analyzed in comparison with those of the depression caused by different forms of stress. It turned out that mice exhibited depressive-like behavior after FMT. Then, PCR array analysis was performed on the hippocampus of the depressed mice to identify differentially expressed genes (DEGs). The KEGG analysis revealed that the pathways of depression induced by gut microbes are closely associated with immuno-inflammation. To determine the pathogenic pathways of physiological stress and psychological stress-induced depression, raw data was extracted from several databases and KEGG analysis was performed. The results from the analysis revealed that the mechanisms of depression induced by physiological and psychological stress are closely related to the regulation of neurotransmitters and energy metabolism. Interestingly, the immunoinflammatory response was distinct across different etiologies that induced depression. The findings showed that gut microbiota dysbiosis-induced depression was mainly associated with adaptive immunity, while physiological stress-induced depression was more linked to innate immunity. This study compared the pathogenesis of depression caused by gut microbiota dysbiosis, and physiological and psychological stress. We explored new intervention methods for depression and laid the foundation for precise treatment.
Collapse
Affiliation(s)
- Xueyi Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; School of Basic Medical Sciences, Department of Pathology, Chongqing Medical University, Chongqing 400016, China
| | - Xiaolong Mo
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yangdong Zhang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Dian He
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Rui Xiao
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; School of Basic Medical Sciences, Department of Pathology, Chongqing Medical University, Chongqing 400016, China
| | - Qisheng Cheng
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Haiyang Wang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Lanxiang Liu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing 402160, China
| | - Wen-Wen Li
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; School of Basic Medical Sciences, Department of Pathology, Chongqing Medical University, Chongqing 400016, China; Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing 400016, China; Department of Pathology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Peng Xie
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing 402160, China.
| |
Collapse
|
15
|
Fang Y, Pan H, Zhu H, Wang H, Ye M, Ren J, Peng J, Li J, Lu X, Huang C. Intranasal LAG3 antibody infusion induces a rapid antidepressant effect via the hippocampal ERK1/2-BDNF signaling pathway in chronically stressed mice. Neuropharmacology 2024; 259:110118. [PMID: 39153731 DOI: 10.1016/j.neuropharm.2024.110118] [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: 06/04/2024] [Revised: 08/05/2024] [Accepted: 08/14/2024] [Indexed: 08/19/2024]
Abstract
The decline of microglia in the dentate gyrus is a new phenomenon that may explain the pathogenesis of depression, and reversing this decline has an antidepressant effect. The development of strategies that restore the function of dentate gyrus microglia in under stressful conditions is becoming a new focus. Lymphocyte-activating gene-3 (LAG3) is an immune checkpoint expressed by immune cells including microglia. One of its functions is to suppress the expansion of immune cells. In a recent study, chronic systemic administration of a LAG3 antibody that readily penetrates the brain was reported to reverse chronic stress-induced hippocampal microglia decline and depression-like behaviors. We showed here that a single intranasal infusion of a LAG3 antibody (In-LAG3 Ab) reversed chronic unpredictable stress (CUS)-induced depression-like behaviors in a dose-dependent manner, which was accompanied by an increase in brain-derived neurotrophic factor (BDNF) in the dentate gyrus. Infusion of an anti-BDNF antibody into the dentate gyrus, construction of knock-in mice with the BDNF Val68Met allele, or treatment with the BDNF receptor antagonist K252a abolished the antidepressant effect of In-LAG3 Ab. Activation of extracellular signal-regulated kinase1/2 (ERK1/2) is required for the reversal effect of In-LAG3 Ab on CUS-induced depression-like behaviors and BDNF decrease in the dentate gyrus. Moreover, both inhibition and depletion of microglia prevented the reversal effect of In-LAG3 Ab on CUS-induced depression-like behaviors and impairment of ERK1/2-BDNF signaling in the dentate gyrus. These results suggest that In-LAG3 Ab exhibits an antidepressant effect through microglia-mediated activation of ERK1/2 and synthesis of BDNF in the dentate gyrus.
Collapse
Affiliation(s)
- Yunli Fang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Hainan Pan
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Haojie Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Hanxiao Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Minxiu Ye
- Department of Pharmacy, Kunshan Hospital of Traditional Chinese Medicine, #388 Zuchongzhi South Road, Kunshan, Suzhou, 215300, China
| | - Jie Ren
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Jie Peng
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Jinxin Li
- Department of Pharmacy, Changzhou Geriatric Hospital Affiliated to Soochow University, Changzhou No. 7 People's Hospital, #288 Yanling East Road, Changzhou 223000, Jiangsu, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China.
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China.
| |
Collapse
|
16
|
Lu Y, Yu X, Wang Z, Kong L, Jiang Z, Shang R, Zhong X, Lv S, Zhang G, Gao H, Yang N. Microbiota-gut-brain axis: Natural antidepressants molecular mechanism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:156012. [PMID: 39260135 DOI: 10.1016/j.phymed.2024.156012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/23/2024] [Accepted: 08/29/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Major depressive disorder (MDD) is a severe mental health condition characterized by persistent depression, impaired cognition, and reduced activity. Increasing evidence suggests that gut microbiota (GM) imbalance is closely linked to the emergence and advancement of MDD, highlighting the potential significance of regulating the "Microbiota-Gut-Brain" (MGB) axis to impact the development of MDD. Natural products (NPs), characterized by broad biological activities, low toxicity, and multi-target characteristics, offer unique advantages in antidepressant treatment by regulating MGB axis. PURPOSE This review was aimed to explore the intricate relationship between the GM and the brain, as well as host responses, and investigated the mechanisms underlying the MGB axis in MDD development. It also explored the pharmacological mechanisms by which NPs modulate MGB axis to exert antidepressant effects and addressed current research limitations. Additionally, it proposed new strategies for future preclinical and clinical applications in the MDD domain. METHODS To study the effects and mechanism by which NPs exert antidepressant effects through mediating the MGB axis, data were collected from Web of Science, PubMed, ScienceDirect from initial establishment to March 2024. NPs were classified and summarized by their mechanisms of action. RESULTS NPs, such as flavonoids,alkaloids,polysaccharides,saponins, terpenoids, can treat MDD by regulating the MGB axis. Its mechanism includes balancing GM, regulating metabolites and neurotransmitters such as SCAFs, 5-HT, BDNF, inhibiting neuroinflammation, improving neural plasticity, and increasing neurogenesis. CONCLUSIONS NPs display good antidepressant effects, and have potential value for clinical application in the prevention and treatment of MDD by regulating the MGB axis. However, in-depth study of the mechanisms by which antidepressant medications affect MGB axis will also require considerable effort in clinical and preclinical research, which is essential for the development of effective antidepressant treatments.
Collapse
Affiliation(s)
- Yitong Lu
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Xiaowen Yu
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Department of Neurology, Affiliated Hospital of shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Zhongling Wang
- Department of Neurology, Affiliated Hospital of shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Linghui Kong
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Zhenyuan Jiang
- Department of Neurology, Affiliated Hospital of shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Ruirui Shang
- College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xia Zhong
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing 100191, China
| | - Shimeng Lv
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Guangheng Zhang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Haonan Gao
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Ni Yang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| |
Collapse
|
17
|
Gordon JA, Dzirasa K, Petzschner FH. The neuroscience of mental illness: Building toward the future. Cell 2024; 187:5858-5870. [PMID: 39423804 PMCID: PMC11490687 DOI: 10.1016/j.cell.2024.09.028] [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: 09/13/2024] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 10/21/2024]
Abstract
Mental illnesses arise from dysfunction in the brain. Although numerous extraneural factors influence these illnesses, ultimately, it is the science of the brain that will lead to novel therapies. Meanwhile, our understanding of this complex organ is incomplete, leading to the oft-repeated trope that neuroscience has yet to make significant contributions to the care of individuals with mental illnesses. This review seeks to counter this narrative, using specific examples of how neuroscientific advances have contributed to progress in mental health care in the past and how current achievements set the stage for further progress in the future.
Collapse
Affiliation(s)
- Joshua A Gordon
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; New York State Psychiatric Institute, New York, NY, USA.
| | - Kafui Dzirasa
- Departments of Psychiatry and Behavioral Sciences, Neurology, and Biomedical Engineering, Duke University Medical Center, Durham, NC, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | | |
Collapse
|
18
|
Paganin W, Signorini S. Inflammatory biomarkers in depression: scoping review. BJPsych Open 2024; 10:e165. [PMID: 39343996 PMCID: PMC11536280 DOI: 10.1192/bjo.2024.787] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/18/2024] [Accepted: 07/11/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Inflammation is increasingly recognised as a fundamental component of the pathophysiology of major depressive disorder (MDD), with a variety of inflammatory biomarkers playing pivotal roles. These markers are closely linked to both the severity of symptoms and the responsiveness to treatments in MDD. AIMS This scoping review aims to explore the scientific literature investigating the complex relationships between inflammatory biomarkers and depression, by identifying new studies and critical issues in current research. METHOD Following the PRISMA Extension for Scoping Reviews guidelines, we systematically searched databases including PubMed, Scopus, PsycINFO, Open Grey and Cochrane Library. Our search focused on articles published from 1 January 2020 to 1 May 2024. We included studies evaluating inflammatory biomarkers in adult patients with MDD, utilising observational and randomised controlled trial designs, and review studies. RESULTS Our analysis examined 44 studies on the complex interplay between inflammation and its multiple effects on MDD. Significant associations between specific inflammatory biomarkers and depression severity were found, requiring cautious interpretation. We also highlight several methodological limitations in the current studies, which warrant caution in directly applying these findings to clinical practice. However, identified methodologies show potential for using these biomarkers as diagnostic tools or therapeutic targets, including anti-inflammatory interventions. CONCLUSIONS The findings emphasise the need for sophisticated, integrative research to understand inflammation's role in MDD. Future studies should identify specific biomarker panels for diagnosing depression and bridging peripheral biomarker measurements with central neuroinflammatory processes, leading to better diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Walter Paganin
- School of Neuroscience, University of Tor Vergata, Italy
| | | |
Collapse
|
19
|
Wang L, Liu L, Liu X, Yang L. The association between neutrophil percentage-to-albumin ratio (NPAR) and depression among US adults: a cross-sectional study. Sci Rep 2024; 14:21880. [PMID: 39300155 DOI: 10.1038/s41598-024-71488-y] [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: 06/12/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024] Open
Abstract
Inflammation plays an important role in depression, and the neutrophil-to-albumin ratio (NPAR) is a cost-effective and readily available novel biomarker of inflammation. The association between NPAR and depression is unclear; therefore, to assess the relationship between NPAR and depression, we conducted a cross-sectional study of 33,768 participants ≥ 18 years of age from the 2005-2018 NHANES database. NPAR was calculated as Neutrophil percentage (in total WBC count) (%) × 100/Albumin (g/dL). Multivariate logistic regression models were used to test the independent association between NPAR and depression, adjusting for demographic factors, education, smoking status, alcohol consumption, hypertension, diabetes mellitus, body mass index, the ratio of income to poverty, and history of cardiovascular disease. Results showed that NPAR was significantly and positively associated with depression. When NPAR were analyzed as a categorical variable, there was a 20% increase in the prevalence of depression in the quartile with the highest NPAR compared to the quartile with the lowest NPAR (OR 1.20[95% CI 1.06, 1.36]). Smoothed curve fitting and threshold effect analyses also showed a positive association between NPAR and depression, with an inflection point for threshold and saturation effects of 12.65. NPAR was positively associated with the likelihood of developing depression when NPAR > 12.65 (OR 1.06[95% CI 1.04, 1.09]). The results of subgroup analyses and interaction tests indicated that smoking status had a significant effect on the relationship between NPAR and depression (P < 0.05). Our study reveals a positive association between NPAR levels and depression, suggesting that higher NPAR levels are associated with an increased likelihood of developing depression.
Collapse
Affiliation(s)
- Lina Wang
- School of Nursing and Rehabilitation, Shandong University, Jinan, 250000, Shandong, China
| | - Li Liu
- Department of Nursing, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250000, China
| | - Xiaojun Liu
- School of Nursing and Rehabilitation, Shandong University, Jinan, 250000, Shandong, China
| | - Lijuan Yang
- Department of Nursing, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250000, China.
| |
Collapse
|
20
|
Yin Y, Ju T, Zeng D, Duan F, Zhu Y, Liu J, Li Y, Lu W. "Inflamed" depression: A review of the interactions between depression and inflammation and current anti-inflammatory strategies for depression. Pharmacol Res 2024; 207:107322. [PMID: 39038630 DOI: 10.1016/j.phrs.2024.107322] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/13/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Depression is a common mental disorder, the effective treatment of which remains a challenging issue worldwide. The clinical pathogenesis of depression has been deeply explored, leading to the formulation of various pathogenic hypotheses. Among these, the monoamine neurotransmitter hypothesis holds a prominent position, yet it has significant limitations as more than one-third of patients do not respond to conventional treatments targeting monoamine transmission disturbances. Over the past few decades, a growing body of research has highlighted the link between inflammation and depression as a potential key factor in the pathophysiology of depression. In this review, we first summarize the relationship between inflammation and depression, with a focus on the pathophysiological changes mediated by inflammation in depression. The mechanisms linking inflammation to depression as well as multiple anti-inflammatory strategies are also discussed, and their efficacy and safety are assessed. This review broadens the perspective on specific aspects of using anti-inflammatory strategies for treating depression, laying the groundwork for advancing precision medicine for individuals suffering from "inflamed" depression.
Collapse
Affiliation(s)
- Yishu Yin
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Ting Ju
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Deyong Zeng
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Fangyuan Duan
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Yuanbing Zhu
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Junlian Liu
- China Astronaut Research and Training Center, Beijing 100094, China
| | - Yongzhi Li
- China Astronaut Research and Training Center, Beijing 100094, China.
| | - Weihong Lu
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China.
| |
Collapse
|
21
|
Zhang PF, You WY, Gao YJ, Wu XB. Activation of pyramidal neurons in the infralimbic cortex alleviates LPS-induced depressive-like behavior in mice. Brain Res Bull 2024; 214:111008. [PMID: 38866373 DOI: 10.1016/j.brainresbull.2024.111008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/23/2024] [Accepted: 06/07/2024] [Indexed: 06/14/2024]
Abstract
The infralimbic (IL) cortex dysfunction has been implicated in major depressive disorder (MDD), yet the precise cellular and molecular mechanisms remain poorly understood. In this study, we investigated the role of layer V pyramidal neurons in a mouse model of MDD induced by repeated lipopolysaccharide (LPS) administration. Our results demonstrate that three days of systemic LPS administration induced depressive-like behavior and upregulated mRNA levels of interleukin-1β (IL-1β), tumor necrosis factor-alpha (TNF-α), and transforming growth factor-β (TGF-β) in the IL cortex. Electrophysiological recordings revealed a significant decrease in the intrinsic excitability of layer V pyramidal neurons in the IL following systemic LPS exposure. Importantly, chemogenetic activation of IL pyramidal neurons ameliorated LPS-induced depressive-like behavior. Additionally, LPS administration significantly increased microglial activity in the IL, as evidenced by a greater number of Ionized calcium binding adaptor molecule-1 (IBA-1)-positive cells. Morphometric analysis further unveiled enlarged soma, decreased branch numbers, and shorter branch lengths of microglial cells in the IL cortex following LPS exposure. Moreover, the activation of pyramidal neurons by clozapine-N-oxide increased the microglia branch length but did not change branch number or cytosolic area. These results collectively suggest that targeted activation of pyramidal neurons in the IL cortex mitigates microglial response and ameliorates depressive-like behaviors induced by systemic LPS administration. Therefore, our findings offer potential therapeutic targets for the development of interventions aimed at alleviating depressive symptoms by modulating IL cortical circuitry and microglial activity.
Collapse
Affiliation(s)
- Peng-Fei Zhang
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu 226019, China
| | - Wen-Yong You
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu 226019, China
| | - Yong-Jing Gao
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu 226019, China.
| | - Xiao-Bo Wu
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu 226019, China.
| |
Collapse
|
22
|
Liu Y, Li C, Ren H, Han K, Wang X, Zang S, Zhao G. The relationship of peripheral blood cell inflammatory biomarkers and psychological stress in unmedicated major depressive disorder. J Psychiatr Res 2024; 176:155-162. [PMID: 38865865 DOI: 10.1016/j.jpsychires.2024.06.013] [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: 12/22/2023] [Revised: 05/07/2024] [Accepted: 06/08/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Recent research has explored the linkage between major depressive disorder (MDD) and inflammation, especially via altered peripheral blood immune markers. However, the relationship between several novel leukocyte-derived ratios (LDR) and psychological stress in MDD remains uncertain. This study aimed to explore the relationship between LDR, clinical characteristics, recent life events, and childhood maltreatment in MDD patients. METHODS A cross-sectional case-control study was conducted involving 59 healthy controls (HC) and 50 unmedicated MDD patients. Subjects underwent psychological assessments and peripheral blood measurements. LDR assessed in this study included neutrophil-to-lymphocyte ratio (NLR), derived NLR (dNLR), monocyte-to-lymphocyte ratio (MLR), platelet-to-lymphocyte ratio (PLR), white blood cell-to-mean platelet volume ratio (WMR), systemic immune inflammation index (SII), multiplication of neutrophil and monocyte counts (MNM), and systemic inflammation response index (SIRI). RESULTS MDD patients displayed significant alterations in WMR, PLR, and MNM compared to HC, as well as correlations between several LDR and various clinical features (duration of untreated psychosis and dNLR, the nine-item Patient Health Questionnaire and PLR, the 7-item Generalized Anxiety Disorder Questionnaire and SIRI (NLR and dNLR). There was a significant difference in the comparison of WMR in first-episode patients than in recurrent patients. Analyses further revealed an association between Life Event Scale total scores and NLR (dNLR). No correlation was found between Childhood Trauma Questionnaire total (or subscale) scores and LDR. Additionally, WMR and dNLR presented potential predictive value for distinguishing between MDD and HC. CONCLUSION The study concludes that MDD and some clinical features are associated with alterations in some peripheral blood LDR. These findings emphasize the potential role of peripheral blood LDR in the pathogenesis and clinical heterogeneity of MDD.
Collapse
Affiliation(s)
- Yigang Liu
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Cuicui Li
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Honghong Ren
- Department of Psychology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Ke Han
- Department of Rehabilitation, Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan, Shandong, China
| | - Ximing Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shuqi Zang
- Department of Rehabilitation, Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan, Shandong, China
| | - Guoqing Zhao
- Department of Psychology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
23
|
Wang JY, Ren P, Cui LY, Duan JY, Chen HL, Zeng ZR, Li YF. Astrocyte-specific activation of sigma-1 receptors in mPFC mediates the faster onset antidepressant effect by inhibiting NF-κB-induced neuroinflammation. Brain Behav Immun 2024; 120:256-274. [PMID: 38852761 DOI: 10.1016/j.bbi.2024.06.008] [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: 04/01/2024] [Revised: 05/29/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024] Open
Abstract
Major depressive disorder (MDD) is a global health burden characterized by persistent low mood, deprivation of pleasure, recurrent thoughts of death, and physical and cognitive deficits. The current understanding of the pathophysiology of MDD is lacking, resulting in few rapid and effective antidepressant therapies. Recent studies have pointed to the sigma-1 (σ-1) receptor as a potential rapid antidepressant target; σ-1 agonists have shown promise in a variety of preclinical depression models. Hypidone hydrochloride (YL-0919), an independently developed antidepressant by our institute with faster onset of action and low rate of side effects, has recently emerged as a highly selective σ-1 receptor agonist; however, its underlying astrocyte-specific mechanism is unknown. In this study, we investigated the effect of YL-0919 treatment on gene expression in the prefrontal cortex of depressive-like mice by single-cell RNA sequencing. Furthermore, we knocked down σ-1 receptors on astrocytes in the medial prefrontal cortex of mice to explore the effects of YL-0919 on depressive-like behavior and neuroinflammation in mice. Our results demonstrated that astrocyte-specific knockdown of σ-1 receptor resulted in depressive-like behavior in mice, which was reversed by YL-0919 administration. In addition, astrocytic σ-1 receptor deficiency led to activation of the NF-κB inflammatory pathway, and crosstalk between reactive astrocytes and activated microglia amplified neuroinflammation, exacerbating stress-induced neuronal apoptosis. Furthermore, the depressive-like behavior induced by astrocyte-specific knockdown of the σ-1 receptor was improved by a selective NF-κB inhibitor, JSH-23, in mice. Our study not only reaffirms the σ-1 receptor as a key target of the faster antidepressant effect of YL-0919, but also contributes to the development of astrocytic σ-1 receptor-based novel drugs.
Collapse
Affiliation(s)
- Jing-Ya Wang
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Peng Ren
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China.
| | - Lin-Yu Cui
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, 100850, China
| | - Jing-Yao Duan
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, 100850, China
| | - Hong-Lei Chen
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Zhi-Rui Zeng
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, 561113, China
| | - Yun-Feng Li
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China; Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, 100850, China.
| |
Collapse
|
24
|
Debler RA, Gallegos PL, Ojeda AC, Perttula AM, Lucio A, Chapkin RS, Safe S, Eitan S. TCDD (2,3,7,8-tetrachlorodibenzo-p-dioxin) induces depression-like phenotype. Neurotoxicology 2024; 103:71-77. [PMID: 38838945 PMCID: PMC11288769 DOI: 10.1016/j.neuro.2024.05.010] [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: 03/03/2024] [Revised: 05/27/2024] [Accepted: 05/31/2024] [Indexed: 06/07/2024]
Abstract
The etiology of major depressive disorder (MDD) remains poorly understood. Our previous studies suggest a role for the aryl hydrocarbon receptor (AhR) in depression. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a toxic environmental contaminant, with a high AhR binding affinity, and an established benchmark for assessing AhR activity. Therefore, this study examined the effect of TCDD on depression-like behaviors. Female mice were fed standard chow or a high-fat diet (HFD) for 11 weeks, and their weight was recorded. Subsequently, they were tested for baseline sucrose preference and splash test grooming. Then, TCDD (0.1 µg/kg/day) or vehicle was administered orally for 28 days, and mice were examined for their sucrose preference and performances in the splash test, forced swim test (FST), and Morris water maze (MWM) task. TCDD significantly decreased sucrose preference, increased FST immobility time, and decreased groom time in chow-fed mice. HFD itself significantly reduced sucrose preference. However, TCDD significantly increased FST immobility time and decreased groom time in HFD-fed mice. A small decrease in bodyweight was observed only at the fourth week of daily TCDD administration in chow-fed mice, and no significant effects of TCDD on bodyweights were observed in HFD-fed mice. TCDD did not have a significant effect on spatial learning in the MWM. Thus, this study demonstrated that TCDD induces a depression-like state, and the effects were not due to gross lethal toxicity. This study further suggests that more studies should examine a possible role for AhR and AhR-active environmental pollutants in precipitating or worsening MDD.
Collapse
Affiliation(s)
- Roanna A Debler
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Paula L Gallegos
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Alexandra C Ojeda
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Andrea M Perttula
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Ashley Lucio
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Robert S Chapkin
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX 77843-4466, USA
| | - Shoshana Eitan
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA.
| |
Collapse
|
25
|
Chen N, Xu X, Guo Y, Zhao M, Li Y, Zhou T, Zhang X, Gao J, Zhu F, Guo C, Shi Y, Wang Q, Wu W, Zhang L, Li Y. Brain Short-Chain Fatty Acids Induce ACSS2 to Ameliorate Depressive-Like Behavior via PPARγ-TPH2 Axis. RESEARCH (WASHINGTON, D.C.) 2024; 7:0400. [PMID: 38939042 PMCID: PMC11210491 DOI: 10.34133/research.0400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/10/2024] [Indexed: 06/29/2024]
Abstract
Short-chain fatty acids (SCFAs) have been increasingly evidenced to be important bioactive metabolites of the gut microbiota and transducers in controlling diverse psychiatric or neurological disorders via the microbiota-gut-brain axis. However, the precise mechanism by which brain SCFAs extert multiple beneficial effects is not completely understood. Our previous research has demonstrated that the acetyl-coenzyme A synthetase short-chain family member 2 (ACSS2) is a novel target of the rapid and long-lasting antidepressant responses. Here, we show that micromolar SCFAs significantly augment both total cellular and nuclear ACSS2 to trigger tryptophan hydroxylase 2 (TPH2) promoter histone acetylation and its transcription in SH-SY5Y cells. In chronic-restraint-stress-induced depression mice, neuronal ACSS2 knockdown by stereotaxic injection of adeno-associated virus in the hippocampus abolished SCFA-mediated improvements in depressive-like behaviors of mice, supporting that ACSS2 is required for SCFA-mediated antidepressant responses. Mechanistically, the peroxisome-proliferator-activated receptor gamma (PPARγ) is identified as a novel partner of ACSS2 to activate TPH2 transcription. Importantly, PPARγ is also responsible for SCFA-mediated antidepressant-like effects via ACSS2-TPH2 axis. To further support brain SCFAs as a therapeutic target for antidepressant effects, d-mannose, which is a naturally present hexose, can significantly reverse the dysbiosis of gut microbiota in the chronic-restraint-stress-exposure mice and augment brain SCFAs to protect against the depressive-like behaviors via ACSS2-PPARγ-TPH2 axis. In summary, brain SCFAs can activate ACSS2-PPARγ-TPH2 axis to play the antidepressive-like effects, and d-mannose is suggested to be an inducer of brain SCFAs in resisting depression.
Collapse
Affiliation(s)
- Nuo Chen
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Xinyi Xu
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Yaxin Guo
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Ming Zhao
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Yubin Li
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Tian Zhou
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Xinyue Zhang
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Jie Gao
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Faliang Zhu
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Chun Guo
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Yongyu Shi
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Qun Wang
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Wenxian Wu
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology,
Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lining Zhang
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| | - Yan Li
- Department of Pathogen Biology, School of Basic Medical Science, Cheeloo College of Medicine,
Shandong University, Jinan, China
| |
Collapse
|
26
|
Shao Y, Chen Y, Wang S, Li C, Sun H, Sun X. Suspected duloxetine-induced restless legs syndrome phenotypic variant: a case report. BMC Psychiatry 2024; 24:349. [PMID: 38730422 PMCID: PMC11088019 DOI: 10.1186/s12888-024-05763-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/13/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Restless arms syndrome (RAS) is the most common variant of restless legs syndrome (RLS), which is easy to be ignored in clinical practice due to the lack of specific diagnostic criteria. When effective therapeutic agents induced RAS and symptoms persisted after briefly observation, clinicians will face the challenge of weighing efficacy against side effects. CASE PRESENTATION A 67-year-old woman was admitted to a geriatric psychiatric ward with depression. Upon admission, the escitalopram dose was reduced from 15 mg to 10 mg per day, and the duloxetine dose was increased from 60 mg to 80 mg per day. The next night before bedtime, she developed itching and creeping sensations deep inside bilateral shoulders and arms, with the urge to move, worsening at rest, and alleviation after hammering. The symptoms persisted when escitalopram was discontinued. A history of RLS was confirmed. Treatment with 40 mg of duloxetine and 0.125 mg of pramipexole significantly improved depression, and the paresthesia disappeared, with no recurrence occurring 6 months after discharge. DISCUSSION AND CONCLUSIONS This case suggests that psychiatrists should pay attention to RLS variants when increasing doses of duloxetine. Long-term improvement can be achieved through dosage reduction combined with dopaminergic drugs instead of immediate discontinuation.
Collapse
Affiliation(s)
- Yan Shao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 51 Huayuan Bei Road, Haidian District, 100191, Beijing, China
| | - Yi Chen
- Ordos Fourth People's Hospital, Ordos, China
| | - Shichang Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 51 Huayuan Bei Road, Haidian District, 100191, Beijing, China
| | - Chaowei Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 51 Huayuan Bei Road, Haidian District, 100191, Beijing, China
| | - Hongqiang Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 51 Huayuan Bei Road, Haidian District, 100191, Beijing, China
| | - Xinyu Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 51 Huayuan Bei Road, Haidian District, 100191, Beijing, China.
| |
Collapse
|
27
|
Toader C, Dobrin N, Costea D, Glavan LA, Covache-Busuioc RA, Dumitrascu DI, Bratu BG, Costin HP, Ciurea AV. Mind, Mood and Microbiota-Gut-Brain Axis in Psychiatric Disorders. Int J Mol Sci 2024; 25:3340. [PMID: 38542314 PMCID: PMC10970241 DOI: 10.3390/ijms25063340] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/09/2024] [Accepted: 03/13/2024] [Indexed: 04/30/2025] Open
Abstract
Psychiatric disorders represent a primary source of disability worldwide, manifesting as disturbances in individuals' cognitive processes, emotional regulation, and behavioral patterns. In the quest to discover novel therapies and expand the boundaries of neuropharmacology, studies from the field have highlighted the gut microbiota's role in modulating these disorders. These alterations may influence the brain's processes through the brain-gut axis, a multifaceted bidirectional system that establishes a connection between the enteric and central nervous systems. Thus, probiotic and prebiotic supplements that are meant to influence overall gut health may play an insightful role in alleviating psychiatric symptoms, such as the cognitive templates of major depressive disorder, anxiety, or schizophrenia. Moreover, the administration of psychotropic drugs has been revealed to induce specific changes in a microbiome's diversity, suggesting their potential utility in combating bacterial infections. This review emphasizes the intricate correlations between psychiatric disorders and the gut microbiota, mentioning the promising approaches in regard to the modulation of probiotic and prebiotic treatments, as well as the antimicrobial effects of psychotropic medication.
Collapse
Affiliation(s)
- Corneliu Toader
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (B.-G.B.); (H.-P.C.); (A.V.C.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Nicolaie Dobrin
- Neurosurgical Clinic, “Prof. Dr. N. Oblu” Emergency Clinical Hospital, 700309 Iași, Romania;
| | - Daniel Costea
- Department of Neurosurgery, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Luca-Andrei Glavan
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (B.-G.B.); (H.-P.C.); (A.V.C.)
| | - Razvan-Adrian Covache-Busuioc
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (B.-G.B.); (H.-P.C.); (A.V.C.)
| | - David-Ioan Dumitrascu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (B.-G.B.); (H.-P.C.); (A.V.C.)
| | - Bogdan-Gabriel Bratu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (B.-G.B.); (H.-P.C.); (A.V.C.)
| | - Horia-Petre Costin
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (B.-G.B.); (H.-P.C.); (A.V.C.)
| | - Alexandru Vlad Ciurea
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (B.-G.B.); (H.-P.C.); (A.V.C.)
- Neurosurgery Department, Sanador Clinical Hospital, 010991 Bucharest, Romania
| |
Collapse
|
28
|
Kouba BR, de Araujo Borba L, Borges de Souza P, Gil-Mohapel J, Rodrigues ALS. Role of Inflammatory Mechanisms in Major Depressive Disorder: From Etiology to Potential Pharmacological Targets. Cells 2024; 13:423. [PMID: 38474387 PMCID: PMC10931285 DOI: 10.3390/cells13050423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
The involvement of central and peripheral inflammation in the pathogenesis and prognosis of major depressive disorder (MDD) has been demonstrated. The increase of pro-inflammatory cytokines (interleukin (IL)-1β, IL-6, IL-18, and TNF-α) in individuals with depression may elicit neuroinflammatory processes and peripheral inflammation, mechanisms that, in turn, can contribute to gut microbiota dysbiosis. Together, neuroinflammation and gut dysbiosis induce alterations in tryptophan metabolism, culminating in decreased serotonin synthesis, impairments in neuroplasticity-related mechanisms, and glutamate-mediated excitotoxicity. This review aims to highlight the inflammatory mechanisms (neuroinflammation, peripheral inflammation, and gut dysbiosis) involved in the pathophysiology of MDD and to explore novel anti-inflammatory therapeutic approaches for this psychiatric disturbance. Several lines of evidence have indicated that in addition to antidepressants, physical exercise, probiotics, and nutraceuticals (agmatine, ascorbic acid, and vitamin D) possess anti-inflammatory effects that may contribute to their antidepressant properties. Further studies are necessary to explore the therapeutic benefits of these alternative therapies for MDD.
Collapse
Affiliation(s)
- Bruna R. Kouba
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis 88040-900, SC, Brazil; (B.R.K.); (L.d.A.B.); (P.B.d.S.)
| | - Laura de Araujo Borba
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis 88040-900, SC, Brazil; (B.R.K.); (L.d.A.B.); (P.B.d.S.)
| | - Pedro Borges de Souza
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis 88040-900, SC, Brazil; (B.R.K.); (L.d.A.B.); (P.B.d.S.)
| | - Joana Gil-Mohapel
- Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Ana Lúcia S. Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis 88040-900, SC, Brazil; (B.R.K.); (L.d.A.B.); (P.B.d.S.)
| |
Collapse
|
29
|
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
|