1
|
Chen Y, Liu Y, Pu J, Gui S, Wang D, Zhong X, Tao W, Chen X, Chen W, Chen X, Qiao R, Li Z, Tao X, Xie P. Treatment response of venlafaxine induced alterations of gut microbiota and metabolites in a mouse model of depression. Metab Brain Dis 2024; 39:1505-1521. [PMID: 39150654 DOI: 10.1007/s11011-024-01403-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 08/05/2024] [Indexed: 08/17/2024]
Abstract
Antidepressants remain the first-line treatment for depression. However, the factors influencing medication response are still unclear. Accumulating evidence implicates an association between alterations in gut microbiota and antidepressant response. Therefore, the aim of this study is to investigate the role of the gut microbiota-brain axis in the treatment response of venlafaxine. After chronic social defeat stress and venlafaxine treatment, mice were divided into responders and non-responders groups. We compared the composition of gut microbiota using 16 S ribosomal RNA sequencing. Meanwhile, we quantified metabolomic alterations in serum and hippocampus, as well as hippocampal neurotransmitter levels using liquid chromatography-mass spectrometry. We found that the abundances of 29 amplicon sequence variants (ASVs) were significantly altered between the responders and non-responders groups. These ASVs belonged to 8 different families, particularly Muribaculaceae. Additionally, we identified 38 and 39 differential metabolites in serum and hippocampus between the responders and non-responders groups, respectively. Lipid, amino acid, and purine metabolisms were enriched in both serum and hippocampus. In hippocampus, the concentrations of tryptophan, phenylalanine, gamma-aminobutyric acid, glutamic acid, and glutamine were increased, while the level of succinic acid was decreased in the responders group, compared with the non-responders group. Our findings suggest that the gut microbiota may play a role in the antidepressant effect of venlafaxine by modulating metabolic processes in the central and peripheral tissues. This provides a novel microbial and metabolic framework for understanding the impact of the gut microbiota-brain axis on antidepressant response.
Collapse
Affiliation(s)
- Yue Chen
- 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, 1 Youyi road, Yuzhong District, Chongqing, 400016, China
| | - Yiyun 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, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi road, Yuzhong District, Chongqing, 400016, China
| | - Juncai Pu
- 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, 1 Youyi road, Yuzhong District, Chongqing, 400016, China
| | - Siwen Gui
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Dongfang Wang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaogang Zhong
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wei Tao
- 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, 1 Youyi road, Yuzhong District, Chongqing, 400016, China
| | - Xiaopeng Chen
- 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, 1 Youyi road, Yuzhong District, Chongqing, 400016, China
| | - Weiyi Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiang Chen
- 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, 1 Youyi road, Yuzhong District, Chongqing, 400016, China
| | - Renjie Qiao
- 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, 1 Youyi road, Yuzhong District, Chongqing, 400016, China
| | - Zhuocan Li
- Psychologic Medicine Science, Chongqing Medical University, Chongqing, China
| | - Xiangkun Tao
- Psychologic Medicine Science, Chongqing Medical University, Chongqing, 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, 1 Youyi road, Yuzhong District, Chongqing, 400016, China.
- Chongqing Institute for Brain and Intelligence, Chongqing, China.
| |
Collapse
|
2
|
Ruiz-Santiago C, Rodríguez-Pinacho CV, Pérez-Sánchez G, Acosta-Cruz E. Effects of selective serotonin reuptake inhibitors on endocrine system (Review). Biomed Rep 2024; 21:128. [PMID: 39070109 PMCID: PMC11273194 DOI: 10.3892/br.2024.1816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/10/2024] [Indexed: 07/30/2024] Open
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are typically prescribed for treating major depressive disorder (MDD) due to their high efficacy. These drugs function by inhibiting the reuptake of serotonin [also termed 5-hydroxytryptamine (5-HT)], which raises the levels of 5-HT in the synaptic cleft, leading to prolonged activation of postsynaptic 5-HT receptors. Despite the therapeutic benefits of SSRIs, this mechanism of action also disturbs the neuroendocrine response. Hypothalamic-pituitary-adrenal (HPA) axis activity is strongly linked to both MDD and the response to antidepressants, owing to the intricate interplay within the serotonergic system, which regulates feeding, water intake, sexual drive, reproduction and circadian rhythms. The aim of the present review was to provide up-to-date evidence for the proposed effects of SSRIs, such as fluoxetine, citalopram, escitalopram, paroxetine, sertraline and fluvoxamine, on the endocrine system. For this purpose, the literature related to the effects of SSRIs on the endocrine system was searched using the PubMed database. According to the available literature, SSRIs may have an adverse effect on glucose metabolism, sexual function and fertility by dysregulating the function of the HPA axis, pancreas and gonads. Therefore, considering that SSRIs are often prescribed for extended periods, it is crucial to monitor the patient closely with particular attention to the function of the endocrine system.
Collapse
Affiliation(s)
- Carolina Ruiz-Santiago
- Department of Biotechnology, Faculty of Chemistry, Universidad Autónoma de Coahuila, Saltillo Coahuila 25280, México
| | | | - Gilberto Pérez-Sánchez
- Laboratory of Psychoimmunology, National Institute of Psychiatry Ramón de la Fuente Muñíz, México City 14370, México
| | - Erika Acosta-Cruz
- Department of Biotechnology, Faculty of Chemistry, Universidad Autónoma de Coahuila, Saltillo Coahuila 25280, México
| |
Collapse
|
3
|
Khan MM, Khan ZA, Khan MA. Metabolic complications of psychotropic medications in psychiatric disorders: Emerging role of de novo lipogenesis and therapeutic consideration. World J Psychiatry 2024; 14:767-783. [PMID: 38984346 PMCID: PMC11230099 DOI: 10.5498/wjp.v14.i6.767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 05/05/2024] [Accepted: 05/23/2024] [Indexed: 06/19/2024] Open
Abstract
Although significant advances have been made in understanding the patho-physiology of psychiatric disorders (PDs), therapeutic advances have not been very convincing. While psychotropic medications can reduce classical symptoms in patients with PDs, their long-term use has been reported to induce or exaggerate various pre-existing metabolic abnormalities including diabetes, obesity and non-alcoholic fatty liver disease (NAFLD). The mechanism(s) underlying these metabolic abnormalities is not clear; however, lipid/fatty acid accumulation due to enhanced de novo lipogenesis (DNL) has been shown to reduce membrane fluidity, increase oxidative stress and inflammation leading to the development of the aforementioned metabolic abnormalities. Intriguingly, emerging evidence suggest that DNL dysregulation and fatty acid accumulation could be the major mechanisms associated with the development of obesity, diabetes and NAFLD after long-term treatment with psychotropic medications in patients with PDs. In support of this, several adjunctive drugs comprising of anti-oxidants and anti-inflammatory agents, that are used in treating PDs in combination with psychotropic medications, have been shown to reduce insulin resistance and development of NAFLD. In conclusion, the above evidence suggests that DNL could be a potential pathological factor associated with various metabolic abnormalities, and a new avenue for translational research and therapeutic drug designing in PDs.
Collapse
Affiliation(s)
- Mohammad M Khan
- Laboratory of Translational Neurology and Molecular Psychiatry, Department of Biotechnology, Era’s Lucknow Medical College and Hospital, and Faculty of Science, Era University, Lucknow 226003, India
| | - Zaw Ali Khan
- Era’s Lucknow Medical College and Hospital, Era University, Lucknow 226003, India
| | - Mohsin Ali Khan
- Era’s Lucknow Medical College and Hospital, Era University, Lucknow 226003, India
| |
Collapse
|
4
|
Sharma AK, Khandelwal R, Wolfrum C. Futile cycles: Emerging utility from apparent futility. Cell Metab 2024; 36:1184-1203. [PMID: 38565147 DOI: 10.1016/j.cmet.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/15/2024] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
Futile cycles are biological phenomena where two opposing biochemical reactions run simultaneously, resulting in a net energy loss without appreciable productivity. Such a state was presumed to be a biological aberration and thus deemed an energy-wasting "futile" cycle. However, multiple pieces of evidence suggest that biological utilities emerge from futile cycles. A few established functions of futile cycles are to control metabolic sensitivity, modulate energy homeostasis, and drive adaptive thermogenesis. Yet, the physiological regulation, implication, and pathological relevance of most futile cycles remain poorly studied. In this review, we highlight the abundance and versatility of futile cycles and propose a classification scheme. We further discuss the energetic implications of various futile cycles and their impact on basal metabolic rate, their bona fide and tentative pathophysiological implications, and putative drug interactions.
Collapse
Affiliation(s)
- Anand Kumar Sharma
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.
| | - Radhika Khandelwal
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Christian Wolfrum
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.
| |
Collapse
|
5
|
Babaaeyan H, Sakhaie N, Sadegzadeh F, Saadati H, Niapour A. Cardiac and hepatic side effects of fluoxetine in male and female adolescent rats. Fundam Clin Pharmacol 2024; 38:320-327. [PMID: 37937375 DOI: 10.1111/fcp.12963] [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: 04/28/2023] [Revised: 09/27/2023] [Accepted: 10/17/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND Fluoxetine (FLX) is widely prescribed as an antidepressant medicine in the juvenile population. OBJECTIVES Although some adverse effects of FLX have been reported in adults, the present study aimed to investigate the side effects of FLX treatment during adolescence on the cardiac and hepatic systems. METHODS Male and female rats were gavaged with FLX (5 mg/kg/day) on postnatal days (PND) 21 to PND 60. Following treatment, blood samples were collected and hepatic enzymes were evaluated. The specimens of the liver and heart of animals were subjected to histopathological assessment. RESULTS Fluoxetine significantly raised serum alanine aminotransferase (ALT) and alkaline phosphatase (ALP) in males, whereas the aspartate aminotransferase (AST) level increased in both male and female animals. In the histopathological study, hepatic plates were more seriously affected, and the sinusoids were irregular in adolescent male rats. Degenerative changes were observed especially in the first and second hepatic zones of FLX-treated male rats. Signs of inflammation and accumulation of lymphoid groups were frequently observed in the portal triad of the hepatic lobules. These alterations were more severe in male livers. Minimum or nearly normal changes were observed in female liver slides. In addition, the histological assessment indicated that treatment with FLX during adolescence also increased the heart's weight and the wall thickness of the right and left ventricles (hypertrophy) in male and especially female animals. CONCLUSION Our findings may provide new insights into the cardiac and hepatic adverse effects of FLX.
Collapse
Affiliation(s)
- Hajar Babaaeyan
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nona Sakhaie
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Farshid Sadegzadeh
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hakimeh Saadati
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali Niapour
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
6
|
Yan JZ, Li GX, Sun SR, Cui LY, Yin YY, Li YF. A rate-limiting step in antidepressants onset: Excitation of glutamatergic pyramidal neurons in medial prefrontal cortex of rodents. Prog Neuropsychopharmacol Biol Psychiatry 2024; 130:110911. [PMID: 38065287 DOI: 10.1016/j.pnpbp.2023.110911] [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: 08/22/2023] [Revised: 11/25/2023] [Accepted: 12/04/2023] [Indexed: 12/25/2023]
Abstract
Although clinical antidepressants have varied mechanisms of action, it remains unclear whether they may have a common mechanism underlying their antidepressant effects. We investigated the behavioral effects of five different antidepressants (differing in target, chemical structure, and rate of onset) and their effects on the firing activities of glutamatergic pyramidal neurons in the medial prefrontal cortex (mPFC) using the forced swimming test (FST) and electrophysiological techniques (in vivo). We employed fiber photometry recordings to validate the effects of antidepressants on the firing activity of pyramidal neurons. Additionally, multichannel electrophysiological recordings were conducted in mice exhibiting depressive-like behaviors induced by chronic restraint stress (CRS) to investigate whether antidepressants exert similar effects on pyramidal neurons in depressed mice. Behavioral tests were utilized for evaluating the depression model. We found that fluoxetine, duloxetine, vilazodone, YL-0919, and ketamine all increase the firing activities of glutamatergic pyramidal neurons (at least 57%) while exerting their initial onset of antidepressant effects. Fiber photometry revealed an increase in the calcium activity of pyramidal neurons in the mPFC at the onset of antidepressant effects. Furthermore, a significant reduction was observed in the firing activity of pyramidal neurons in the mPFC of CRS-exposed mice, which was reversed by antidepressants. Taken together, our findings suggested that five pharmacologically distinct classes of antidepressants share the common ability to increase the firing activity of pyramidal neurons, just different time, which might be a rate-limiting step in antidepressants onset. The study contributes to the body of knowledge of the mechanisms underlying antidepressant effects and paves the way for developing rapid-acting antidepressants.
Collapse
Affiliation(s)
- Jiao-Zhao Yan
- Beijing Institute of Basic Medical Sciences, Beijing, China; Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Guang-Xiang Li
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Si-Rui Sun
- Beijing Ditan Hospital Capital Medical University, Beijing, China
| | - Lin-Yu Cui
- College of Anesthesia, Shanxi Medical University, Shanxi, China
| | - Yong-Yu Yin
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China.
| | - Yun-Feng Li
- Beijing Institute of Basic Medical Sciences, Beijing, China; Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China.
| |
Collapse
|
7
|
Zorkina Y, Ushakova V, Ochneva A, Tsurina A, Abramova O, Savenkova V, Goncharova A, Alekseenko I, Morozova I, Riabinina D, Kostyuk G, Morozova A. Lipids in Psychiatric Disorders: Functional and Potential Diagnostic Role as Blood Biomarkers. Metabolites 2024; 14:80. [PMID: 38392971 PMCID: PMC10890164 DOI: 10.3390/metabo14020080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 02/25/2024] Open
Abstract
Lipids are a crucial component of the human brain, serving important structural and functional roles. They are involved in cell function, myelination of neuronal projections, neurotransmission, neural plasticity, energy metabolism, and neuroinflammation. Despite their significance, the role of lipids in the development of mental disorders has not been well understood. This review focused on the potential use of lipids as blood biomarkers for common mental illnesses, such as major depressive disorder, anxiety disorders, bipolar disorder, and schizophrenia. This review also discussed the impact of commonly used psychiatric medications, such as neuroleptics and antidepressants, on lipid metabolism. The obtained data suggested that lipid biomarkers could be useful for diagnosing psychiatric diseases, but further research is needed to better understand the associations between blood lipids and mental disorders and to identify specific biomarker combinations for each disease.
Collapse
Affiliation(s)
- Yana Zorkina
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (V.U.); (A.O.); (A.T.); (O.A.); (V.S.); (I.M.); (D.R.); (G.K.); (A.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| | - Valeria Ushakova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (V.U.); (A.O.); (A.T.); (O.A.); (V.S.); (I.M.); (D.R.); (G.K.); (A.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| | - Aleksandra Ochneva
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (V.U.); (A.O.); (A.T.); (O.A.); (V.S.); (I.M.); (D.R.); (G.K.); (A.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| | - Anna Tsurina
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (V.U.); (A.O.); (A.T.); (O.A.); (V.S.); (I.M.); (D.R.); (G.K.); (A.M.)
| | - Olga Abramova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (V.U.); (A.O.); (A.T.); (O.A.); (V.S.); (I.M.); (D.R.); (G.K.); (A.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| | - Valeria Savenkova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (V.U.); (A.O.); (A.T.); (O.A.); (V.S.); (I.M.); (D.R.); (G.K.); (A.M.)
| | - Anna Goncharova
- Moscow Center for Healthcare Innovations, 123473 Moscow, Russia;
| | - Irina Alekseenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academi of Science, 142290 Moscow, Russia
- Russia Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, 2, Kurchatov Square, 123182 Moscow, Russia
| | - Irina Morozova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (V.U.); (A.O.); (A.T.); (O.A.); (V.S.); (I.M.); (D.R.); (G.K.); (A.M.)
| | - Daria Riabinina
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (V.U.); (A.O.); (A.T.); (O.A.); (V.S.); (I.M.); (D.R.); (G.K.); (A.M.)
| | - Georgy Kostyuk
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (V.U.); (A.O.); (A.T.); (O.A.); (V.S.); (I.M.); (D.R.); (G.K.); (A.M.)
| | - Anna Morozova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (V.U.); (A.O.); (A.T.); (O.A.); (V.S.); (I.M.); (D.R.); (G.K.); (A.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| |
Collapse
|
8
|
Gamboa-Sánchez C, Becerril-Villanueva E, Alvarez-Herrera S, Leyva-Mascareño G, González-López SL, Estudillo E, Fernández-Molina AE, Elizalde-Contreras JM, Ruiz-May E, Segura-Cabrera A, Jiménez-Genchi J, Pavón L, Zamudio SR, Pérez-Sánchez G. Upregulation of S100A8 in peripheral blood mononuclear cells from patients with depression treated with SSRIs: a pilot study. Proteome Sci 2023; 21:23. [PMID: 38049858 PMCID: PMC10694904 DOI: 10.1186/s12953-023-00224-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/17/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND Major depressive disorder (MDD) affects more than 350 million people worldwide, and there is currently no laboratory test to diagnose it. This pilot study aimed to identify potential biomarkers in peripheral blood mononuclear cells (PBMCs) from MDD patients. METHODS We used tandem mass tagging coupled to synchronous precursor selection (mass spectrometry) to obtain the differential proteomic profile from a pool of PBMCs from MDD patients and healthy subjects, and quantitative PCR to assess gene expression of differentially expressed proteins (DEPs) of our interest. RESULTS We identified 247 proteins, of which 133 had a fold change ≥ 2.0 compared to healthy volunteers. Using pathway enrichment analysis, we found that some processes, such as platelet degranulation, coagulation, and the inflammatory response, are perturbed in MDD patients. The gene-disease association analysis showed that molecular alterations in PBMCs from MDD patients are associated with cerebral ischemia, vascular disease, thrombosis, acute coronary syndrome, and myocardial ischemia, in addition to other conditions such as inflammation and diabetic retinopathy. CONCLUSIONS We confirmed by qRT-PCR that S100A8 is upregulated in PBMCs from MDD patients and thus could be an emerging biomarker of this disorder. This report lays the groundwork for future studies in a broader and more diverse population and contributes to a deeper characterization of MDD.
Collapse
Affiliation(s)
- Concepción Gamboa-Sánchez
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de La Fuente Muñiz, Colonia San Lorenzo Huipulco, Calzada México-Xochimilco 101, Tlalpan, 14370, Ciudad de Mexico, México
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional. Unidad Profesional Adolfo López Mateos, Av. Wilfrido Massieu 399, Nueva Industrial Vallejo, Gustavo A. Madero, 07738, Ciudad de México, México
| | - Enrique Becerril-Villanueva
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de La Fuente Muñiz, Colonia San Lorenzo Huipulco, Calzada México-Xochimilco 101, Tlalpan, 14370, Ciudad de Mexico, México
| | - Samantha Alvarez-Herrera
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de La Fuente Muñiz, Colonia San Lorenzo Huipulco, Calzada México-Xochimilco 101, Tlalpan, 14370, Ciudad de Mexico, México
| | - Gabriela Leyva-Mascareño
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de La Fuente Muñiz, Colonia San Lorenzo Huipulco, Calzada México-Xochimilco 101, Tlalpan, 14370, Ciudad de Mexico, México
| | - Sandra L González-López
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de La Fuente Muñiz, Colonia San Lorenzo Huipulco, Calzada México-Xochimilco 101, Tlalpan, 14370, Ciudad de Mexico, México
| | - Enrique Estudillo
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Av. Insurgentes Sur 3877 Del. Tlalpan, 14269. Col. La Fama., Ciudad de México, México
| | - Alberto E Fernández-Molina
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de La Fuente Muñiz, Colonia San Lorenzo Huipulco, Calzada México-Xochimilco 101, Tlalpan, 14370, Ciudad de Mexico, México
| | - José Miguel Elizalde-Contreras
- Red de Estudios Moleculares Avanzados, Instituto de Ecología A. C, Cluster BioMimic®, Carretera Antigua a Coatepec 351, Congregación El Haya, 91073, Xalapa, Veracruz, México
| | - Eliel Ruiz-May
- Red de Estudios Moleculares Avanzados, Instituto de Ecología A. C, Cluster BioMimic®, Carretera Antigua a Coatepec 351, Congregación El Haya, 91073, Xalapa, Veracruz, México
| | - Aldo Segura-Cabrera
- Red de Estudios Moleculares Avanzados, Instituto de Ecología A. C, Cluster BioMimic®, Carretera Antigua a Coatepec 351, Congregación El Haya, 91073, Xalapa, Veracruz, México
- Genomic Sciences, GSK, Stevenage, UK
| | - Janeth Jiménez-Genchi
- Hospital Psiquiátrico Fray Bernardino Álvarez. Av, Niño Jesús, San Buenaventura 214000, Tlalpan, Ciudad de Mexico, México
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de La Fuente Muñiz, Colonia San Lorenzo Huipulco, Calzada México-Xochimilco 101, Tlalpan, 14370, Ciudad de Mexico, México
| | - Sergio Roberto Zamudio
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional. Unidad Profesional Adolfo López Mateos, Av. Wilfrido Massieu 399, Nueva Industrial Vallejo, Gustavo A. Madero, 07738, Ciudad de México, México.
| | - Gilberto Pérez-Sánchez
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de La Fuente Muñiz, Colonia San Lorenzo Huipulco, Calzada México-Xochimilco 101, Tlalpan, 14370, Ciudad de Mexico, México.
| |
Collapse
|
9
|
Gallant RM, Snyder JM, Ayres JS. Fluoxetine promotes immunometabolic defenses to mediate host-pathogen cooperation during sepsis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.18.567681. [PMID: 38013994 PMCID: PMC10680848 DOI: 10.1101/2023.11.18.567681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are some of the most prescribed drugs in the world. While they are used for their ability to increase serotonergic signaling in the brain, SSRIs are also known to have a broad range of effects beyond the brain, including immune and metabolic effects. Recent studies have demonstrated that SSRIs are protective in animal models and humans against several infections, including sepsis and COVID-19, however the mechanisms underlying this protection are largely unknown. Here we mechanistically link two previously described effects of the SSRI fluoxetine in mediating protection against sepsis. We show that fluoxetine-mediated protection is independent of peripheral serotonin, and instead increases levels of circulating IL-10. IL-10 is necessary for protection from sepsis-induced hypertriglyceridemia and cardiac triglyceride accumulation, allowing for metabolic reprogramming of the heart. Our work reveals a beneficial "off-target" effect of fluoxetine, and reveals a protective immunometabolic defense mechanism with therapeutic potential.
Collapse
Affiliation(s)
- Robert M Gallant
- Molecular and Systems Physiology Lab, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92037, USA
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
- Gene Expression Lab, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jessica M Snyder
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle WA
| | - Janelle S Ayres
- Molecular and Systems Physiology Lab, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
- Gene Expression Lab, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA
- Lead contact
| |
Collapse
|
10
|
del Valle E, Rubio-Sardón N, Menéndez-Pérez C, Martínez-Pinilla E, Navarro A. Apolipoprotein D as a Potential Biomarker in Neuropsychiatric Disorders. Int J Mol Sci 2023; 24:15631. [PMID: 37958618 PMCID: PMC10650001 DOI: 10.3390/ijms242115631] [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: 10/08/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Neuropsychiatric disorders (NDs) are a diverse group of pathologies, including schizophrenia or bipolar disorders, that directly affect the mental and physical health of those who suffer from them, with an incidence that is increasing worldwide. Most NDs result from a complex interaction of multiple genes and environmental factors such as stress or traumatic events, including the recent Coronavirus Disease (COVID-19) pandemic. In addition to diverse clinical presentations, these diseases are heterogeneous in their pathogenesis, brain regions affected, and clinical symptoms, making diagnosis difficult. Therefore, finding new biomarkers is essential for the detection, prognosis, response prediction, and development of new treatments for NDs. Among the most promising candidates is the apolipoprotein D (Apo D), a component of lipoproteins implicated in lipid metabolism. Evidence suggests an increase in Apo D expression in association with aging and in the presence of neuropathological processes. As a part of the cellular neuroprotective defense machinery against oxidative stress and inflammation, changes in Apo D levels have been demonstrated in neuropsychiatric conditions like schizophrenia (SZ) or bipolar disorders (BPD), not only in some brain areas but in corporal fluids, i.e., blood or serum of patients. What is not clear is whether variation in Apo D quantity could be used as an indicator to detect NDs and their progression. This review aims to provide an updated view of the clinical potential of Apo D as a possible biomarker for NDs.
Collapse
Affiliation(s)
- Eva del Valle
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain; (E.d.V.); (N.R.-S.); (C.M.-P.); (A.N.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33006 Oviedo, Spain
| | - Nuria Rubio-Sardón
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain; (E.d.V.); (N.R.-S.); (C.M.-P.); (A.N.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33006 Oviedo, Spain
| | - Carlota Menéndez-Pérez
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain; (E.d.V.); (N.R.-S.); (C.M.-P.); (A.N.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33006 Oviedo, Spain
| | - Eva Martínez-Pinilla
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain; (E.d.V.); (N.R.-S.); (C.M.-P.); (A.N.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33006 Oviedo, Spain
| | - Ana Navarro
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain; (E.d.V.); (N.R.-S.); (C.M.-P.); (A.N.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33006 Oviedo, Spain
| |
Collapse
|
11
|
Arora A, Becker M, Marques C, Oksanen M, Li D, Mastropasqua F, Watts ME, Arora M, Falk A, Daub CO, Lanekoff I, Tammimies K. Screening autism-associated environmental factors in differentiating human neural progenitors with fractional factorial design-based transcriptomics. Sci Rep 2023; 13:10519. [PMID: 37386098 PMCID: PMC10310850 DOI: 10.1038/s41598-023-37488-0] [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: 02/03/2023] [Accepted: 06/22/2023] [Indexed: 07/01/2023] Open
Abstract
Research continues to identify genetic variation, environmental exposures, and their mixtures underlying different diseases and conditions. There is a need for screening methods to understand the molecular outcomes of such factors. Here, we investigate a highly efficient and multiplexable, fractional factorial experimental design (FFED) to study six environmental factors (lead, valproic acid, bisphenol A, ethanol, fluoxetine hydrochloride and zinc deficiency) and four human induced pluripotent stem cell line derived differentiating human neural progenitors. We showcase the FFED coupled with RNA-sequencing to identify the effects of low-grade exposures to these environmental factors and analyse the results in the context of autism spectrum disorder (ASD). We performed this after 5-day exposures on differentiating human neural progenitors accompanied by a layered analytical approach and detected several convergent and divergent, gene and pathway level responses. We revealed significant upregulation of pathways related to synaptic function and lipid metabolism following lead and fluoxetine exposure, respectively. Moreover, fluoxetine exposure elevated several fatty acids when validated using mass spectrometry-based metabolomics. Our study demonstrates that the FFED can be used for multiplexed transcriptomic analyses to detect relevant pathway-level changes in human neural development caused by low-grade environmental risk factors. Future studies will require multiple cell lines with different genetic backgrounds for characterising the effects of environmental exposures in ASD.
Collapse
Affiliation(s)
- Abishek Arora
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, Visionsgatan 4, 171 56, Solna, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden
| | - Martin Becker
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, Visionsgatan 4, 171 56, Solna, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden
| | - Cátia Marques
- Department of Chemistry - BMC, Uppsala University, Uppsala, Sweden
| | - Marika Oksanen
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, Visionsgatan 4, 171 56, Solna, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden
| | - Danyang Li
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, Visionsgatan 4, 171 56, Solna, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden
| | - Francesca Mastropasqua
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, Visionsgatan 4, 171 56, Solna, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden
| | - Michelle Evelyn Watts
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, Visionsgatan 4, 171 56, Solna, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden
| | - Manish Arora
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Lund Stem Cell Center, Division of Neurobiology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Carsten Oliver Daub
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
- Science for Life Laboratory, Stockholm, Sweden
| | - Ingela Lanekoff
- Department of Chemistry - BMC, Uppsala University, Uppsala, Sweden
| | - Kristiina Tammimies
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women's and Children's Health, Karolinska Institutet, BioClinicum J9:30, Visionsgatan 4, 171 56, Solna, Stockholm, Sweden.
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden.
| |
Collapse
|
12
|
Chen L, Ji Y, Li A, Liu B, Shen K, Su R, Ma Z, Zhang W, Wang Q, Zhu Y, Xue W. High-throughput drug screening identifies fluoxetine as a potential therapeutic agent for neuroendocrine prostate cancer. Front Oncol 2023; 13:1085569. [PMID: 36994207 PMCID: PMC10042075 DOI: 10.3389/fonc.2023.1085569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/21/2023] [Indexed: 03/14/2023] Open
Abstract
IntroductionNeuroendocrine prostate cancer (NEPC) is an aggressive subtype of prostate cancer with poor prognosis and resistance to hormone therapy, which has limited therapeutic approaches. Therefore, this study aimed to identify a novel treatment for NEPC and provide evidence of its inhibitory effects.MethodsWe performed a high-throughput drug screening and identified fluoxetine, originally an FDA-approved antidepressant, as candidate therapeutic agent for NEPC. We carried out both in vitro and in vivo experiments to demonstrate the inhibitory effects of fluoxetine on NEPC models and its mechanism in detail.ResultsOur results demonstrated that fluoxetine effectively curbed the neuroendocrine differentiation and inhibited cell viability by targeting the AKT pathway. Preclinical test in NEPC mice model (PBCre4: Ptenf/f; Trp53f/f; Rb1f/f) showed that fluoxetine effectively prolonged the overall survival and reduced the risk of tumor distant metastases.DiscussionThis work repurposed fluoxetine for antitumor application, and supported its clinical development for NEPC therapy, which may provide a promising therapeutic strategy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Qi Wang
- *Correspondence: Qi Wang, ; Yinjie Zhu,
| | | | | |
Collapse
|
13
|
Zhu W, Li W, Jiang J, Wang D, Mao X, Zhang J, Zhang X, Chang J, Yao P, Yang X, Da Costa C, Zhang Y, Yu J, Li H, Li S, Chi X, Li N. Chronic salmon calcitonin exerts an antidepressant effect via modulating the p38 MAPK signaling pathway. Front Mol Neurosci 2023; 16:1071327. [PMID: 36969556 PMCID: PMC10036804 DOI: 10.3389/fnmol.2023.1071327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 02/15/2023] [Indexed: 03/12/2023] Open
Abstract
Depression is a common recurrent psychiatric disorder with a high lifetime prevalence and suicide rate. At present, although several traditional clinical drugs such as fluoxetine and ketamine, are widely used, medications with a high efficiency and reduced side effects are of urgent need. Our group has recently reported that a single administration of salmon calcitonin (sCT) could ameliorate a depressive-like phenotype via the amylin signaling pathway in a mouse model established by chronic restraint stress (CRS). However, the molecular mechanism underlying the antidepressant effect needs to be addressed. In this study, we investigated the antidepressant potential of sCT applied chronically and its underlying mechanism. In addition, using transcriptomics, we found the MAPK signaling pathway was upregulated in the hippocampus of CRS-treated mice. Further phosphorylation levels of ERK/p38/JNK kinases were also enhanced, and sCT treatment was able only to downregulate the phosphorylation level of p38/JNK, with phosphorylated ERK level unaffected. Finally, we found that the antidepressant effect of sCT was blocked by p38 agonists rather than JNK agonists. These results provide a mechanistic explanation of the antidepressant effect of sCT, suggesting its potential for treating the depressive disorder in the clinic.
Collapse
Affiliation(s)
- Wenhui Zhu
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Weifen Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Jian Jiang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Dilong Wang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xinliang Mao
- Perfect Life and Health Institute, Zhongshan, Guangdong, China
| | - Jin Zhang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xunzhi Zhang
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Jinlong Chang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Peijia Yao
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xiuyan Yang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | | | - Ying Zhang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jiezhong Yu
- The Fourth People’s Hospital of Datong City, Datong, China
| | - Huiliang Li
- Wolfson Institute for Biomedical Research, Division of Medicine, Faculty of Medical Sciences, University College London, London, United Kingdom
- China-UK Institute for Frontier Science, Shenzhen, China
- *Correspondence: Huiliang Li,
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
- Shupeng Li,
| | - Xinjin Chi
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Xinjin Chi,
| | - Ningning Li
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- China-UK Institute for Frontier Science, Shenzhen, China
- The Fifth People’s Hospital of Datong City, Datong, China
- Ningning Li,
| |
Collapse
|
14
|
Huang HS, Lin YE, Panyod S, Chen RA, Lin YC, Chai LMX, Hsu CC, Wu WK, Lu KH, Huang YJ, Sheen LY. Anti-depressive-like and cognitive impairment alleviation effects of Gastrodia elata Blume water extract is related to gut microbiome remodeling in ApoE -/- mice exposed to unpredictable chronic mild stress. JOURNAL OF ETHNOPHARMACOLOGY 2023; 302:115872. [PMID: 36343797 DOI: 10.1016/j.jep.2022.115872] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGY RELEVANCE Gastrodia elata Blume (GE) is a traditional Chinese dietary therapy used to treat neurological disorders. Gastrodia elata Blume water extract (WGE) has been shown to ameliorate inflammation and improve social frustration in mice in a chronic social defeat model. However, studies on the anti-depressive-like effects and cognitive impairment alleviation related to the impact of WGE on the gut microbiome of ApoE-/- mice remain elusive. AIM OF THE STUDY The present study aimed to investigate the anti-depressive-like effect and cognitive impairment alleviation and mechanisms of WGE in ApoE-/- mice subjected to unpredictable chronic mild stress (UCMS), as well as its impact on the gut microbiome of the mice. MATERIALS AND METHODS Sixty ApoE-/- mice (6 months old) were randomly grouped into six groups: control, UCMS, WGE groups [5, 10, 20 mL WGE/kg body weight (bw) + UCMS], and a positive group (fluoxetine 20 mg/kg bw + UCMS). After four weeks of the UCMS paradigm, the sucrose preference, novel object recognition, and open field tests were conducted. The neurotransmitters serotonin (5-HT), dopamine (DA) and their metabolites were measured in the prefrontal cortex. Serum was collected to measure corticosterone and amyloid-42 (Aβ-42) levels. Feces were collected, and the gut microbiome was analyzed. RESULTS WGE restored sucrose preference, exploratory behavior, recognition ability, and decreased the levels of serum corticosterone and Aβ-42 in ApoE-/- mice to alleviate depressive-like behavior and cognitive impairment. Furthermore, WGE regulated the monoamine neurotransmitter via reduced the 5-HT and DA turnover rates in the prefrontal cortex. Moreover, WGE elevated the levels of potentially beneficial bacteria such as Bifidobacterium, Akkermansia, Alloprevotella, Defluviitaleaceae_UCG-011, and Bifidobacterium pseudolongum as well as balanced fecal short-chain fatty acids (SCFAs). CONCLUSION WGE demonstrates anti-depressive-like effects, cognitive impairment alleviation, and gut microbiome and metabolite regulation in ApoE-/- mice. Our results support the possibility of developing a functional and complementary medicine to prevent or alleviate depression and cognitive decline using WGE in CVDs patients.
Collapse
Affiliation(s)
- Huai-Syuan Huang
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan.
| | - Yu-En Lin
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan.
| | - Suraphan Panyod
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan.
| | - Rou-An Chen
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan.
| | - Ying-Cheng Lin
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan.
| | | | - Cheng-Chih Hsu
- Department of Chemistry, National Taiwan University, Taipei, Taiwan.
| | - Wei-Kai Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan.
| | - Kuan-Hung Lu
- Institute of Food Safety and Health, National Taiwan University, Taipei, Taiwan; Institute of Environmental and Occupational Health Sciences, National Taiwan University, Taipei, Taiwan.
| | - Yun-Ju Huang
- Department of Biotechnology and Food Technology, Southern Taiwan University of Science and Technology, Tainan, Taiwan.
| | - Lee-Yan Sheen
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan; Center for Food and Biomolecules, National Taiwan University, Taipei, Taiwan; National Center for Food Safety Education and Research, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
15
|
Duarte B, Feijão E, Cruz de Carvalho R, Duarte IA, Marques AP, Maia M, Hertzog J, Matos AR, Cabrita MT, Caçador I, Figueiredo A, Silva MS, Cordeiro C, Fonseca VF. Untargeted Metabolomics Reveals Antidepressant Effects in a Marine Photosynthetic Organism: The Diatom Phaeodactylum tricornutum as a Case Study. BIOLOGY 2022; 11:1770. [PMID: 36552278 PMCID: PMC9775013 DOI: 10.3390/biology11121770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022]
Abstract
The increased use of antidepressants, along with their increased occurrence in aquatic environments, is of concern for marine organisms. Although these pharmaceutical compounds have been shown to negatively affect marine diatoms, their mode of action in these non-target, single-cell phototrophic organisms is yet unknown. Using a Fourier-transform ion cyclotron-resonance mass spectrometer (FT-ICR-MS) we evaluated the effects of fluoxetine in the metabolomics of the model diatom Phaeodactylum tricornutum, as well as the potential use of the identified metabolites as exposure biomarkers. Diatom growth was severely impaired after fluoxetine exposure, particularly in the highest dose tested, along with a down-regulation of photosynthetic and carbohydrate metabolisms. Notably, several mechanisms that are normally down-regulated by fluoxetine in mammal organisms were also down-regulated in diatoms (e.g., glycerolipid metabolism, phosphatidylinositol signalling pathway, vitamin metabolism, terpenoid backbone biosynthesis and serotonin remobilization metabolism). Additionally, the present work also identified a set of potential biomarkers of fluoxetine exposure that were up-regulated with increasing fluoxetine exposure concentration and are of high metabolic significance following the disclosed mode of action, reinforcing the use of metabolomics approaches in ecotoxicology.
Collapse
Affiliation(s)
- Bernardo Duarte
- MARE—Marine and Environmental Sciences Centre & ARNET—Aquatic Research Network Associated Laboratory, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
- Departamento de Biologia Vegetal, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| | - Eduardo Feijão
- MARE—Marine and Environmental Sciences Centre & ARNET—Aquatic Research Network Associated Laboratory, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| | - Ricardo Cruz de Carvalho
- MARE—Marine and Environmental Sciences Centre & ARNET—Aquatic Research Network Associated Laboratory, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
- cE3c, Centre for Ecology, Evolution and Environmental Changes, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, Edifício C2, Piso 5, 1749-016 Lisbon, Portugal
| | - Irina A. Duarte
- MARE—Marine and Environmental Sciences Centre & ARNET—Aquatic Research Network Associated Laboratory, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| | - Ana Patrícia Marques
- MARE—Marine and Environmental Sciences Centre & ARNET—Aquatic Research Network Associated Laboratory, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
- Laboratório de FT-ICR e Espectrometria de Massa Estrutural, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| | - Marisa Maia
- Université de Lorraine, LCP-A2MC, F-57000 Metz, France
| | | | - Ana Rita Matos
- Departamento de Biologia Vegetal, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
- BioISI—Biosystems and Integrative Sciences Institute, Plant Functional Genomics Group, Departamento de Biologia Vegetal, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| | - Maria Teresa Cabrita
- Centro de Estudos Geográficos (CEG), Instituto de Geografia e Ordenamento do Território (IGOT), Universidade de Lisboa, Rua Branca Edmée Marques, 1600-276 Lisbon, Portugal
- Associated Laboratory Terra, 1349-017 Lisbon, Portugal
| | - Isabel Caçador
- MARE—Marine and Environmental Sciences Centre & ARNET—Aquatic Research Network Associated Laboratory, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
- Departamento de Biologia Vegetal, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| | - Andreia Figueiredo
- Departamento de Biologia Vegetal, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
- BioISI—Biosystems and Integrative Sciences Institute, Plant Functional Genomics Group, Departamento de Biologia Vegetal, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| | - Marta Sousa Silva
- MARE—Marine and Environmental Sciences Centre & ARNET—Aquatic Research Network Associated Laboratory, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
- Laboratório de FT-ICR e Espectrometria de Massa Estrutural, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| | - Carlos Cordeiro
- MARE—Marine and Environmental Sciences Centre & ARNET—Aquatic Research Network Associated Laboratory, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
- Laboratório de FT-ICR e Espectrometria de Massa Estrutural, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| | - Vanessa F. Fonseca
- MARE—Marine and Environmental Sciences Centre & ARNET—Aquatic Research Network Associated Laboratory, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
- Departamento de Biologia Animal, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| |
Collapse
|
16
|
Abstract
Obsessive-compulsive disorder (OCD) has a bidirectional relationship with metabolic disorders. The purposes of this review are to decipher the links between OCD and metabolic disorders and to explore the etiological mechanism of OCD in metabolism, which may aid in early identification of and tailored interventions for OCD and metabolic disorders.
Collapse
|
17
|
Is depression the missing link between inflammatory mediators and cancer? Pharmacol Ther 2022; 240:108293. [PMID: 36216210 DOI: 10.1016/j.pharmthera.2022.108293] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/14/2022] [Accepted: 09/26/2022] [Indexed: 11/05/2022]
Abstract
Patients with cancer are at greater risk of developing depression in comparison to the general population and this is associated with serious adverse effects, such as poorer quality of life, worse prognosis and higher mortality. Although the relationship between depression and cancer is now well established, a common underlying pathophysiological mechanism between the two conditions is yet to be elucidated. Existing theories of depression, based on monoamine neurotransmitter system dysfunction, are insufficient as explanations of the disorder. Recent advances have implicated neuroinflammatory mechanisms in the etiology of depression and it has been demonstrated that inflammation at a peripheral level may be mirrored centrally in astrocytes and microglia serving to promote chronic levels of inflammation in the brain. Three major routes to depression in cancer in which proinflammatory mediators are implicated, seem likely. Activation of the kynurenine pathway involving cytokines, increases tryptophan catabolism, resulting in diminished levels of serotonin which is widely acknowledged as being the hallmark of depression. It also results in neurotoxic effects on brain regions thought to be involved in the evolution of major depression. Proinflammatory mediators also play a crucial role in impairing regulatory glucocorticoid mediated feedback of the hypothalamic-pituitary-adrenal axis, which is activated by stress and considered to be involved in both depression and cancer. The third route is via the glutamatergic pathway, whereby glutamate excitotoxicity may lead to depression associated with cancer. A better understanding of the mechanisms underlying these dysregulated and other newly emerging pathways may provide a rationale for therapeutic targeting, serving to improve the care of cancer patients.
Collapse
|
18
|
Cai L, Tao Q, Li W, Zhu X, Cui C. The anti-anxiety/depression effect of a combined complex of casein hydrolysate and γ-aminobutyric acid on C57BL/6 mice. Front Nutr 2022; 9:971853. [PMID: 36245498 PMCID: PMC9554304 DOI: 10.3389/fnut.2022.971853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/12/2022] [Indexed: 12/02/2022] Open
Abstract
In view of a series of adverse side effects of drugs for anxiety/depression on the market at present, it is imminent to extract and develop novel anti-anxiety and depression drugs from plants and proteins (like casein hydrolysate) as adjuncts or substitutes for existing anti-anxiety and depression drugs. Consequently, this study investigated the improvement of the anxiety/depression function by the compound of casein hydrolysate and γ-aminobutyric acid (GABA) (casein hydrolysate: GABA = 4:1; CCHAA) on mice induced by chronic restraint stress-corticosterone injection. Animal experiments revealed that oral gavage administration of CCHAA significantly reversed the anxiety/depression-like behaviors. Compared to the model control group, body weights were increased after treatment with CCHAA groups [1.5, 0.75 mg/(g⋅d)]. As a diagnostic index of anxiety and depression, we assessed GABA and 5-HT levels in response to CCHAA ingestion. The GABA and 5-HT levels were increasingly enhanced by the CCHAA diet. In addition, histopathological changes in the hippocampus CA3 region of the anxious/depressed mice were also alleviated after the treatment with the CCHAA. Thus, the casein hydrolysate and GABA formula diets may induce beneficial effects on the mice with anxiety/depression.
Collapse
Affiliation(s)
- Lei Cai
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Qian Tao
- Infinitus (China) Co., Ltd., Guangzhou, China
| | - Wenzhi Li
- Infinitus (China) Co., Ltd., Guangzhou, China
| | - Xiping Zhu
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, China
- *Correspondence: Xiping Zhu,
| | - Chun Cui
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Guangdong Weiwei Biotechnology Co., Ltd., Guangzhou, China
- Chun Cui,
| |
Collapse
|
19
|
Zhang YM, Ye LY, Li TY, Guo F, Guo F, Li Y, Li YF. New monoamine antidepressant, hypidone hydrochloride (YL-0919), enhances the excitability of medial prefrontal cortex in mice via a neural disinhibition mechanism. Acta Pharmacol Sin 2022; 43:1699-1709. [PMID: 34811511 PMCID: PMC9253340 DOI: 10.1038/s41401-021-00807-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 10/28/2021] [Indexed: 12/28/2022] Open
Abstract
Hypidone hydrochloride (YL-0919) is a novel antidepressant in clinical phase II trial. Previous studies show that YL-0919 is a selective 5-HT (serotonin) reuptake inhibitor, 5-HT1A receptor partial agonist, and 5-HT6 receptor agonist, which exerts antidepressant effects in various animal models, but its effects on neural function remain unclear. Medial prefrontal cortex (mPFC), a highly evolved brain region, controls highest order cognitive functions and emotion regulation. In this study we investigated the effects of YL-0919 on the mPFC function, including the changes in neuronal activities using electrophysiological recordings. Extracellular recording (in vivo) showed that chronic administration of YL-0919 significantly increased the spontaneous discharges of mPFC neurons. In mouse mPFC slices, whole-cell recording revealed that perfusion of YL-0919 significantly increased the frequency of sEPSCs, but decreased the frequency of sIPSCs. Then we conducted whole-cell recording in mPFC slices of GAD67-GFP transgenic mice, and demonstrated that YL-0919 significantly inhibited the excitability of GABAergic neurons. In contrast, it did not alter the excitability of pyramidal neurons in mPFC slices of normal mice. Moreover, the inhibition of GABAergic neurons by YL-0919 was prevented by pre-treatment with 5-HT1A receptor antagonist WAY 100635. Finally, chronic administration of YL-0919 significantly increased the phosphorylation levels of mTOR and GSK-3β in the mPFC as compared with vehicle. Taken together, our results demonstrate that YL-0919 enhances the excitability of mPFC via a disinhibition mechanism to fulfill its rapid antidepressant neural mechanism, which was accomplished by 5-HT1A receptor-mediated inhibition of inhibitory GABAergic interneurons.
Collapse
Affiliation(s)
- Yong-mei Zhang
- grid.419093.60000 0004 0619 8396CAS Key Laboratory of Receptor Research, Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Lu-yu Ye
- grid.419093.60000 0004 0619 8396CAS Key Laboratory of Receptor Research, Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Tian-yu Li
- grid.419093.60000 0004 0619 8396CAS Key Laboratory of Receptor Research, Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Fan Guo
- grid.419093.60000 0004 0619 8396CAS Key Laboratory of Receptor Research, Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Fei Guo
- CAS Key Laboratory of Receptor Research, Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Yang Li
- CAS Key Laboratory of Receptor Research, Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Yun-feng Li
- grid.410740.60000 0004 1803 4911Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, 100850 China
| |
Collapse
|
20
|
Beigi T, Safi A, Satvati M, Kalantari-Hesari A, Ahmadi R, Meshkibaf MH. Protective role of ellagic acid and taurine against fluoxetine induced hepatotoxic effects on biochemical and oxidative stress parameters, histopathological changes, and gene expressions of IL-1β, NF-κB, and TNF-α in male Wistar rats. Life Sci 2022; 304:120679. [PMID: 35662648 DOI: 10.1016/j.lfs.2022.120679] [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: 03/04/2022] [Revised: 05/23/2022] [Accepted: 05/28/2022] [Indexed: 11/27/2022]
Abstract
PURPOSES Hepatic bioactivation of fluoxetine (FXN) could increase free radicals' generation provoking hepatotoxicity. Therefore, the protective effects of ellagic acid (EA) and taurine (TAU) treatments against fluoxetine-induced liver damage in rats were examined. MATERIALS AND METHODS Sixty four male Wistar rats were randomly assigned to 8 groups (n = 8). Group (1) Control, group (2) FXN, group (3) FXN + EA, group (4) FXN + TAU, group (5) FXN + EA + TAU, group (6) EA, group (7) TAU, and group (8) EA + TAU. Then, the serum and tissue parameters of the oxidative stress were examined. KEY FINDINGS FXN significantly raised serum MDA, protein carbonyl, lipid profile, ALT, AST, ALP, total bilirubin, serum IL-1β; and gene expressions of IL-1β, NF-κB, and TNF-α. Moreover, it significantly decreased HDL-C, ferric reducing antioxidant power (FRAP), catalase activity, vitamin C, and SOD activity in the liver compared to group 1. When compared to group 2, EA and TAU treatment dramatically increased antioxidant capacity and lowered hepatotoxic biochemical markers and cellular inflammation. Results also showed a protective effect of treatment against oxidative damage caused by hepatocytes' cytoarchitecture. SIGNIFICANCE Our study concluded the beneficial effects of EA and TAU on FXN-induced hepatotoxicity. These effects were derived from free radical scavenging properties and the anti-inflammatory effects related to IL-1β, NF-κB, and TNF-α gene expression inhibition.
Collapse
Affiliation(s)
- Tayebeh Beigi
- Department of Clinical Biochemistry, Fasa University of Medical Sciences, Fasa, Iran
| | - Amir Safi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mahdi Satvati
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Ali Kalantari-Hesari
- Department of Pathobiology, Faculty of Veterinary Science, Bu-Ali Sina University, Hamedan, Iran
| | - Reza Ahmadi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | | |
Collapse
|
21
|
Heindel JJ, Howard S, Agay-Shay K, Arrebola JP, Audouze K, Babin PJ, Barouki R, Bansal A, Blanc E, Cave MC, Chatterjee S, Chevalier N, Choudhury M, Collier D, Connolly L, Coumoul X, Garruti G, Gilbertson M, Hoepner LA, Holloway AC, Howell G, Kassotis CD, Kay MK, Kim MJ, Lagadic-Gossmann D, Langouet S, Legrand A, Li Z, Le Mentec H, Lind L, Monica Lind P, Lustig RH, Martin-Chouly C, Munic Kos V, Podechard N, Roepke TA, Sargis RM, Starling A, Tomlinson CR, Touma C, Vondracek J, Vom Saal F, Blumberg B. Obesity II: Establishing causal links between chemical exposures and obesity. Biochem Pharmacol 2022; 199:115015. [PMID: 35395240 PMCID: PMC9124454 DOI: 10.1016/j.bcp.2022.115015] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 02/06/2023]
Abstract
Obesity is a multifactorial disease with both genetic and environmental components. The prevailing view is that obesity results from an imbalance between energy intake and expenditure caused by overeating and insufficient exercise. We describe another environmental element that can alter the balance between energy intake and energy expenditure: obesogens. Obesogens are a subset of environmental chemicals that act as endocrine disruptors affecting metabolic endpoints. The obesogen hypothesis posits that exposure to endocrine disruptors and other chemicals can alter the development and function of the adipose tissue, liver, pancreas, gastrointestinal tract, and brain, thus changing the set point for control of metabolism. Obesogens can determine how much food is needed to maintain homeostasis and thereby increase the susceptibility to obesity. The most sensitive time for obesogen action is in utero and early childhood, in part via epigenetic programming that can be transmitted to future generations. This review explores the evidence supporting the obesogen hypothesis and highlights knowledge gaps that have prevented widespread acceptance as a contributor to the obesity pandemic. Critically, the obesogen hypothesis changes the narrative from curing obesity to preventing obesity.
Collapse
Affiliation(s)
- Jerrold J Heindel
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, USA.
| | - Sarah Howard
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, USA
| | - Keren Agay-Shay
- Health and Environment Research (HER) Lab, The Azrieli Faculty of Medicine, Bar Ilan University, Israel
| | - Juan P Arrebola
- Department of Preventive Medicine and Public Health University of Granada, Granada, Spain
| | - Karine Audouze
- Department of Systems Biology and Bioinformatics, University of Paris, INSERM, T3S, Paris France
| | - Patrick J Babin
- Department of Life and Health Sciences, University of Bordeaux, INSERM, Pessac France
| | - Robert Barouki
- Department of Biochemistry, University of Paris, INSERM, T3S, 75006 Paris, France
| | - Amita Bansal
- College of Health & Medicine, Australian National University, Canberra, Australia
| | - Etienne Blanc
- Department of Biochemistry, University of Paris, INSERM, T3S, 75006 Paris, France
| | - Matthew C Cave
- Division of Gastroenterology, Hepatology and Nutrition, University of Louisville, Louisville, KY 40402, USA
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, University of South Carolina, Columbia, SC 29208, USA
| | - Nicolas Chevalier
- Obstetrics and Gynecology, University of Cote d'Azur, Cote d'Azur, France
| | - Mahua Choudhury
- College of Pharmacy, Texas A&M University, College Station, TX 77843, USA
| | - David Collier
- Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Lisa Connolly
- The Institute for Global Food Security, School of Biological Sciences, Queen's University, Belfast, Northern Ireland, UK
| | - Xavier Coumoul
- Department of Biochemistry, University of Paris, INSERM, T3S, 75006 Paris, France
| | - Gabriella Garruti
- Department of Endocrinology, University of Bari "Aldo Moro," Bari, Italy
| | - Michael Gilbertson
- Occupational and Environmental Health Research Group, University of Stirling, Stirling, Scotland
| | - Lori A Hoepner
- Department of Environmental and Occupational Health Sciences, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Alison C Holloway
- McMaster University, Department of Obstetrics and Gynecology, Hamilton, Ontario, CA, USA
| | - George Howell
- Center for Environmental Health Sciences, Mississippi State University, Mississippi State, MS 39762, USA
| | - Christopher D Kassotis
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, USA
| | - Mathew K Kay
- College of Pharmacy, Texas A&M University, College Station, TX 77843, USA
| | - Min Ji Kim
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | | | - Sophie Langouet
- Univ Rennes, INSERM EHESP, IRSET UMR_5S 1085, 35000 Rennes, France
| | - Antoine Legrand
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Zhuorui Li
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Helene Le Mentec
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Lars Lind
- Clinical Epidemiology, Department of Medical Sciences, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - P Monica Lind
- Occupational and Environmental Medicine, Department of Medical Sciences, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Robert H Lustig
- Division of Endocrinology, Department of Pediatrics, University of California San Francisco, CA 94143, USA
| | | | - Vesna Munic Kos
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Normand Podechard
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Troy A Roepke
- Department of Animal Science, School of Environmental and Biological Science, Rutgers University, New Brunswick, NJ 08901, USA
| | - Robert M Sargis
- Division of Endocrinology, Diabetes and Metabolism, The University of Illinois at Chicago, Chicago, Il 60612, USA
| | - Anne Starling
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Craig R Tomlinson
- Norris Cotton Cancer Center, Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Charbel Touma
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Jan Vondracek
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Frederick Vom Saal
- Division of Biological Sciences, The University of Missouri, Columbia, MO 65211, USA
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
22
|
Effects of pharmacological treatment on metabolomic alterations in animal models of depression. Transl Psychiatry 2022; 12:175. [PMID: 35487889 PMCID: PMC9055046 DOI: 10.1038/s41398-022-01947-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 04/17/2022] [Accepted: 04/21/2022] [Indexed: 12/16/2022] Open
Abstract
Numerous studies have investigated metabolite alterations resulting from pharmacological treatment in depression models although few quantitative studies explored metabolites exhibiting constant alterations. This study aimed to identify consistently dysregulated metabolites across such studies using a knowledgebase-driven approach. This study was based on 157 studies that identified an assembly of 2757 differential metabolites in the brain, blood, urine, liver, and feces samples of depression models with pharmacological medication. The use of a vote-counting approach to identify consistently upregulated and downregulated metabolites showed that serotonin, dopamine, norepinephrine, gamma-aminobutyric acid, anandamide, tryptophan, hypoxanthine, and 3-methoxytyramine were upregulated in the brain, while quinolinic acid, glutamic acid, 5-hydroxyindoleacetic acid, myo-inositol, lactic acid, and the kynurenine/tryptophan ratio were downregulated. Circulating levels of trimethylamine N-oxide, isoleucine, leucine, tryptophan, creatine, serotonin, valine, betaine, and low-density lipoprotein were elevated. In contrast, levels of alpha-D-glucose, lactic acid, N-acetyl glycoprotein, glutamine, beta-D-glucose, corticosterone, alanine, phenylacetylglycine, glycine, high-density lipoprotein, arachidonic acid, myo-inositol, allantoin, and taurine were decreased. Moreover, 12 metabolites in urine and nine metabolites in the liver were dysregulated after treatment. Pharmacological treatment also increased fecal levels of butyric acid, acetic acid, propionic acid, and isovaleric acid. Collectively, metabolite disturbances induced by depression were reversed by pharmacological treatment. Pharmacological medication reversed the reduction of brain neurotransmitters caused by depression, modulated disturbance of the tryptophan-kynurenine pathway and inflammatory activation, and alleviated abnormalities of amino acid metabolism, energy metabolism, lipid metabolism, and gut microbiota-derived metabolites.
Collapse
|
23
|
Yang R, Wang L, Cao S, Chen M, Wu CJ, Silva F, Shen MJ, Chen JD, Tang MM, Liu BL. Sex difference in lipid levels in first-diagnosed drug-naïve depression patients: A case-control and 12-weeks follow-up study. World J Biol Psychiatry 2022; 23:228-235. [PMID: 34320901 DOI: 10.1080/15622975.2021.1961500] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
AIM Patients with depression have a high prevalence of developing dyslipidemia. In this study, we aim to investigate the difference of serum lipids, including total cholesterol (TCH), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), and triglycerides (TG), between the depressed patients and healthy controls. Sex differences in lipids and their psychological correlations were also included. METHODS The study included 56 healthy controls (males/females = 26/30) and 110 first-diagnosed drug-naïve outpatients (males/females = 35/75). A total of 42 patients (males/females = 14/28) were followed for 3 months. RESULTS A significant difference was found in TCH and LDL-C among healthy control and patients. Interestingly, female patients with first-diagnosed, drug-naïve depression had lower atherogenic indices than male patients. After 3 months of antidepressants therapy, female patients exhibited detrimental changes in serum lipids, namely increased TG and atherogenic index. Moreover, correlation analysis showed significant correlations between changes of depression inventory (HAMD and BDI) score and serum lipids (TCH, HDL-C) in depressed patients. CONCLUSION We found that dyslipidemia was more common in female patients with depression during therapy with antidepressants. Moreover, the altered serum lipids and atherogenic index might be a hallmark of female patients. Further investigation of sex differences in lipid metabolism of depression is warranted.
Collapse
Affiliation(s)
- Rui Yang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Institute for Rational and Safe Medication Practices, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lu Wang
- Mental Health Institute of the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Song Cao
- Mental Health Institute of the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ming Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Institute for Rational and Safe Medication Practices, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Chu-Jun Wu
- Mental Health Institute of the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Floyd Silva
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Man-Jun Shen
- Mental Health Institute of the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jin-Dong Chen
- Mental Health Institute of the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mi-Mi Tang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Institute for Rational and Safe Medication Practices, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bi-Lian Liu
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
24
|
Ito N, Maruko A, Oshima K, Yoshida M, Honma K, Sugiyama C, Nagai T, Kobayashi Y, Odaguchi H, Okada N. Kampo formulas alleviate aging-related emotional disturbances and neuroinflammation in male senescence-accelerated mouse prone 8 mice. Aging (Albany NY) 2022; 14:109-142. [PMID: 34979499 PMCID: PMC8791223 DOI: 10.18632/aging.203811] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/20/2021] [Indexed: 11/25/2022]
Abstract
Aging-induced neuroinflammation, also known as neuroinflammaging, plays a pivotal role in emotional disturbances, including depression and anxiety, in older individuals, thereby leading to cognitive dysfunction. Although numerous studies have focused on therapeutic strategies for cognitive impairment in older individuals, little research has been performed on treating its preceding emotional disturbances. Here, we examined whether Kampo formulas (kososan [KS], nobiletin-rich kososan [NKS], and hachimijiogan [HJG]) can ameliorate aging-induced emotional disturbances and neuroinflammation in mice. The depression-like behaviors observed in SAMP8 mice, relative to normally aging SAMR1 mice, were significantly prevented by treatment with Kampo formulas for 13 weeks. Western blot analysis revealed that hippocampal neuroinflammation was significantly abrogated by Kampo formulas. KS and NKS also significantly attenuated the hippocampal neuroinflammatory priming induced by lipopolysaccharide (LPS, 0.33 mg/kg, i.p.) challenge in SAMP8 mice. Hippocampal IL-1β, IL-6, and MCP-1 levels were significantly decreased in NKS-treated SAMP8 mice. KS and NKS showed significantly reduced tau accumulation in the brains of SAMP8 mice. RNA-sequencing revealed that each Kampo formula led to unique dynamics of hippocampal gene expression and appeared to abrogate hippocampal inflammatory responses. HJG significantly blocked the LPS-induced increase in serum IL-6 and MCP-1. These results suggest that Kampo formulas would be useful for treating aging-induced depression, in part by regulating neuroinflammatory pathways. This finding may pave the way for the development of therapeutic strategies for aging-related emotional disturbances, which may contribute to the prevention of cognitive dysfunction in older individuals.
Collapse
Affiliation(s)
- Naoki Ito
- Oriental Medicine Research Center, Kitasato University, Tokyo 108-8642, Japan
| | - Akiko Maruko
- Laboratory of Genomics for Health and Longevity, School of Pharmacy, Kitasato University, Sagamihara, Kanagawa 252-0373, Japan
| | - Kenshiro Oshima
- Laboratory of Genomics for Health and Longevity, School of Pharmacy, Kitasato University, Sagamihara, Kanagawa 252-0373, Japan
| | - Masaaki Yoshida
- Research Laboratory, Kotaro Pharmaceutical Co., Ltd., Hakusan, Ishikawa 920-0201, Japan
| | - Kengo Honma
- Graduate School of Infection Control Sciences, Kitasato University, Tokyo 108-8642, Japan
| | - Chika Sugiyama
- Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa 252-0373, Japan
| | - Takayuki Nagai
- Oriental Medicine Research Center, Kitasato University, Tokyo 108-8642, Japan.,Graduate School of Infection Control Sciences, Kitasato University, Tokyo 108-8642, Japan.,Laboratory of Biochemical Pharmacology for Phytomedicines, Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo 108-8642, Japan
| | - Yoshinori Kobayashi
- Oriental Medicine Research Center, Kitasato University, Tokyo 108-8642, Japan.,Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa 252-0373, Japan
| | - Hiroshi Odaguchi
- Oriental Medicine Research Center, Kitasato University, Tokyo 108-8642, Japan.,Department of Pharmacognosy, School of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8642, Japan
| | - Norihiro Okada
- Laboratory of Genomics for Health and Longevity, School of Pharmacy, Kitasato University, Sagamihara, Kanagawa 252-0373, Japan
| |
Collapse
|
25
|
Dimoula A, Fotellis D, Aivalioti E, Delialis D, Polissidis A, Patras R, Kokras N, Stamatelopoulos K. Off-Target Effects of Antidepressants on Vascular Function and Structure. Biomedicines 2021; 10:biomedicines10010056. [PMID: 35052735 PMCID: PMC8773150 DOI: 10.3390/biomedicines10010056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 12/22/2022] Open
Abstract
Depression emerges as a risk factor for cardiovascular disease, and it is thought that successful antidepressant treatment may reduce such a risk. Therefore, antidepressant treatment embodies a potential preventive measure to reduce cardiovascular events in patients with depression. Accumulating evidence indicates that antidepressants have off-target effects on vascular dysfunction and in the early stages of atherosclerosis, which form the basis for cardiovascular disease (CVD) pathogenesis. In this context, we performed a thorough review of the evidence pertaining to the effects of different classes of antidepressant medications on hemodynamic and early atherosclerosis markers. The preclinical and clinical evidence reviewed revealed a preponderance of studies assessing selective serotonin reuptake inhibitors (SSRI), whereas other classes of antidepressants are less well-studied. Sufficient evidence supports a beneficial effect of SSRIs on vascular inflammation, endothelial function, arterial stiffening, and possibly delaying carotid atherosclerosis. In clinical studies, dissecting the hypothesized direct beneficial antidepressant effect of SSRIs on endothelial health from the global improvement upon remission of depression has proven to be difficult. However, preclinical studies armed with appropriate control groups provide evidence of molecular mechanisms linked to endothelial function that are indeed modulated by antidepressants. This suggests at least a partial direct action on vascular integrity. Further research on endothelial markers should focus on the effect of antidepressants on treatment responders versus non-responders in order to better ascertain the possible beneficial vascular effects of antidepressants, irrespective of the underlying course of depression.
Collapse
Affiliation(s)
- Anna Dimoula
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 80 Vas. Sofias Str., 11528 Athens, Greece; (A.D.); (D.F.); (E.A.); (D.D.); (R.P.)
| | - Dimitrios Fotellis
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 80 Vas. Sofias Str., 11528 Athens, Greece; (A.D.); (D.F.); (E.A.); (D.D.); (R.P.)
| | - Evmorfia Aivalioti
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 80 Vas. Sofias Str., 11528 Athens, Greece; (A.D.); (D.F.); (E.A.); (D.D.); (R.P.)
| | - Dimitrios Delialis
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 80 Vas. Sofias Str., 11528 Athens, Greece; (A.D.); (D.F.); (E.A.); (D.D.); (R.P.)
| | - Alexia Polissidis
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.P.); (N.K.)
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), 4 Soranou Efesiou St., 11527 Athens, Greece
- First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Raphael Patras
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 80 Vas. Sofias Str., 11528 Athens, Greece; (A.D.); (D.F.); (E.A.); (D.D.); (R.P.)
| | - Nikolaos Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.P.); (N.K.)
- First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 80 Vas. Sofias Str., 11528 Athens, Greece; (A.D.); (D.F.); (E.A.); (D.D.); (R.P.)
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK
- Correspondence:
| |
Collapse
|
26
|
Ayyash A, Holloway AC. Fluoxetine-induced hepatic lipid accumulation is mediated by prostaglandin endoperoxide synthase 1 and is linked to elevated 15-deoxy-Δ 12,14 PGJ 2. J Appl Toxicol 2021; 42:1004-1015. [PMID: 34897744 DOI: 10.1002/jat.4272] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/17/2021] [Indexed: 12/11/2022]
Abstract
Major depressive disorder and other neuropsychiatric disorders are often managed with long-term use of antidepressant medication. Fluoxetine, an SSRI antidepressant, is widely used as a first-line treatment for neuropsychiatric disorders. However, fluoxetine has also been shown to increase the risk of metabolic diseases such as non-alcoholic fatty liver disease. Fluoxetine has been shown to increase hepatic lipid accumulation in vivo and in vitro. In addition, fluoxetine has been shown to alter the production of prostaglandins which have also been implicated in the development of non-alcoholic fatty liver disease. The goal of this study was to assess the effect of fluoxetine exposure on the prostaglandin biosynthetic pathway and lipid accumulation in a hepatic cell line (H4-II-E-C3 cells). Fluoxetine treatment increased mRNA expression of prostaglandin biosynthetic enzymes (Ptgs1, Ptgs2, and Ptgds), PPAR gamma (Pparg), and PPAR gamma downstream targets involved in fatty acid uptake (Cd36, Fatp2, and Fatp5) as well as production of 15-deoxy-Δ12,14 PGJ2 a PPAR gamma ligand. The effects of fluoxetine to induce lipid accumulation were attenuated with a PTGS1 specific inhibitor (SC-560), whereas inhibition of PTGS2 had no effect. Moreover, SC-560 attenuated 15-deoxy-Δ12,14 PGJ2 production and expression of PPAR gamma downstream target genes. Taken together these results suggest that fluoxetine-induced lipid abnormalities appear to be mediated via PTGS1 and its downstream product 15d-PGJ2 and suggest a novel therapeutic target to prevent some of the adverse effects of fluoxetine treatment.
Collapse
Affiliation(s)
- Ahmed Ayyash
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| | - Alison C Holloway
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
27
|
Hu Y, Wang Y, Chen C, Yang W, Zhu W, Wang Y, Liu P. A randomized, placebo-controlled, double-blind study on the effects of SZL on patients with mild to moderate depressive disorder with comparison to fluoxetine. JOURNAL OF ETHNOPHARMACOLOGY 2021; 281:114549. [PMID: 34438029 DOI: 10.1016/j.jep.2021.114549] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Kaixinsan (KXS) decoction, as an herbal formula, was used to treat the diseases, such as insomnia, amnesia, emotional disorders in ancient china. It has been demonstrated to be active in various animal models resembling human depression with multitarget effects. However, effective verification on the clinical application of KXS is still lacking. Supplements in this knowledge field are urgently needed. AIM OF THE STUDY This very first study evaluated the efficacy and tolerability of ShenZhiLing (SZL) tablets (KXS preparation), compared with fluoxetine (FLX, positive comparator), in patients with mild to moderate depressive disorder. MATERIALS AND METHODS In this randomized, double-blind, parallel-group study, 156 patients with mild to moderate depression without taken any antidepressants in the past 6 months or 4 continuous weeks were randomized to receive either 3.2 g/d SZL plus 20 mg/d FLX placebo (SZL group) or 20 mg/d FLX plus 3.2 g/d SZL placebo (FLX group), for 8 weeks. Their clinical presentations and some metabolic indexes were assessed during the 8 weeks' visiting period. RESULTS Patients in SZL group showed a statistically significant improvement after 8 weeks of treatment in HAM-D17 score (18.79±2.09 to 4.43±4.71, p<0.001) and self-rating depression scale (SDS) score (58.49±8.89 to 39.84±12.09, p<0.001), but not in N-back total respond time (1145.55±608.26 to 1128.47±387.49, p>0.05). In addition, no significant difference at 8 weeks of treatment was found between SZL and FLX groups in SDS score (39.84±12.09 vs. 36.63±12.44) and N-back respond time (1128.47±387.49 vs. 1089.43±352.08) as well as reduction of HAM-D17 score (14.79±4.88 vs. 15.24±4.29) (p>0.05 for all). However, the serum APOB, APOC3 and ALB levels and LDL-C/HDL-C ratio decreased significantly in patients after SZL treatment, while only APOB/APOA1 ratio decreased significantly in FLX group. Other metabolic indexes did not alter significantly after treated with SZL or FLX. CONCLUSION The efficacy and safety profile of SZL are comparable to that of fluoxetine in patients with mild to moderate depression. The beneficial effect of SZL is probably associated with improvement of lipid metabolic balance.
Collapse
Affiliation(s)
- Yuan Hu
- Department of Clinical Pharmacology, Medical Supplier Center, Chinese PLA General Hospital, Beijing, 100853, China; Chinese PLA Medical School, Beijing, 100853, China.
| | - Yichen Wang
- Department of Clinical Pharmacology, Medical Supplier Center, Chinese PLA General Hospital, Beijing, 100853, China; Chinese PLA Medical School, Beijing, 100853, China.
| | - Chao Chen
- Department of Clinical Pharmacology, Medical Supplier Center, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Wenshan Yang
- Department of Clinical Pharmacology, Medical Supplier Center, Chinese PLA General Hospital, Beijing, 100853, China; Chinese PLA Medical School, Beijing, 100853, China.
| | - Weiyu Zhu
- Department of Clinical Pharmacology, Medical Supplier Center, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Yuanbo Wang
- Department of Clinical Pharmacology, Medical Supplier Center, Chinese PLA General Hospital, Beijing, 100853, China; Chinese PLA Medical School, Beijing, 100853, China.
| | - Ping Liu
- Department of Clinical Pharmacology, Medical Supplier Center, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
28
|
Park J, Jeong W, Yun C, Kim H, Oh CM. Serotonergic Regulation of Hepatic Energy Metabolism. Endocrinol Metab (Seoul) 2021; 36:1151-1160. [PMID: 34911172 PMCID: PMC8743581 DOI: 10.3803/enm.2021.1331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 11/25/2021] [Indexed: 01/01/2023] Open
Abstract
The liver is a vital organ that regulates systemic energy metabolism and many physiological functions. Nonalcoholic fatty liver disease (NAFLD) is the commonest cause of chronic liver disease and end-stage liver failure. NAFLD is primarily caused by metabolic disruption of lipid and glucose homeostasis. Serotonin (5-hydroxytryptamine [5-HT]) is a biogenic amine with several functions in both the central and peripheral systems. 5-HT functions as a neurotransmitter in the brain and a hormone in peripheral tissues to regulate systemic energy homeostasis. Several recent studies have proposed various roles of 5-HT in hepatic metabolism and inflammation using tissue-specific knockout mice and 5-HT-receptor agonists/antagonists. This review compiles the most recent research on the relationship between 5-HT and hepatic metabolism, and the role of 5-HT signaling as a potential therapeutic target in NAFLD.
Collapse
Affiliation(s)
- Jiwon Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju,
Korea
| | - Wooju Jeong
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju,
Korea
| | - Chahyeon Yun
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju,
Korea
| | - Hail Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon,
Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju,
Korea
| |
Collapse
|
29
|
Luo H, Jiang ZL, Ren Y. Therapy Management of Metabolic Disorder Comorbidity With Depression. Front Psychol 2021; 12:683320. [PMID: 34408704 PMCID: PMC8366060 DOI: 10.3389/fpsyg.2021.683320] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 06/29/2021] [Indexed: 11/29/2022] Open
Abstract
Depression is a common disease that seriously endangers the physical and mental health of human beings, and it often coexists with other metabolic disorders such as diabetes and cancer. There have been endless reports on the mechanism, prevention, and cure of comorbidity because of its high incidence and poor prognosis and the increased burden on the family and society. There may be a specific comorbid basis and causal relationship between depression and metabolic diseases. Depression in patients with metabolic disorders can be effectively alleviated through psychotherapy and medication. The timely and effective treatment of depression can significantly improve the quality of life of patients with metabolic disorders, reduce their psychological burden, and promote the effective treatment of metabolic diseases. This study reorganized the research progress on the management of metabolic disorder comorbidity with depression.
Collapse
Affiliation(s)
- Hua Luo
- Department of Orthopedics, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Zheng-Li Jiang
- Department of Pharmacy, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Yu Ren
- Department of Pharmacy, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| |
Collapse
|
30
|
Tkachev A, Stekolshchikova E, Bobrovskiy DM, Anikanov N, Ogurtsova P, Park DI, Horn AKE, Petrova D, Khrameeva E, Golub MS, Turck CW, Khaitovich P. Long-Term Fluoxetine Administration Causes Substantial Lipidome Alteration of the Juvenile Macaque Brain. Int J Mol Sci 2021; 22:ijms22158089. [PMID: 34360852 PMCID: PMC8348031 DOI: 10.3390/ijms22158089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/13/2022] Open
Abstract
Fluoxetine is an antidepressant commonly prescribed not only to adults but also to children for the treatment of depression, obsessive-compulsive disorder, and neurodevelopmental disorders. The adverse effects of the long-term treatment reported in some patients, especially in younger individuals, call for a detailed investigation of molecular alterations induced by fluoxetine treatment. Two-year fluoxetine administration to juvenile macaques revealed effects on impulsivity, sleep, social interaction, and peripheral metabolites. Here, we built upon this work by assessing residual effects of fluoxetine administration on the expression of genes and abundance of lipids and polar metabolites in the prelimbic cortex of 10 treated and 11 control macaques representing two monoamine oxidase A (MAOA) genotypes. Analysis of 8871 mRNA transcripts, 3608 lipids, and 1829 polar metabolites revealed substantial alterations of the brain lipid content, including significant abundance changes of 106 lipid features, accompanied by subtle changes in gene expression. Lipid alterations in the drug-treated animals were most evident for polyunsaturated fatty acids (PUFAs). A decrease in PUFAs levels was observed in all quantified lipid classes excluding sphingolipids, which do not usually contain PUFAs, suggesting systemic changes in fatty acid metabolism. Furthermore, the residual effect of the drug on lipid abundances was more pronounced in macaques carrying the MAOA-L genotype, mirroring reported behavioral effects of the treatment. We speculate that a decrease in PUFAs may be associated with adverse effects in depressive patients and could potentially account for the variation in individual response to fluoxetine in young people.
Collapse
Affiliation(s)
- Anna Tkachev
- V. Zelman Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia; (A.T.); (E.S.); (N.A.); (P.O.); (D.P.)
| | - Elena Stekolshchikova
- V. Zelman Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia; (A.T.); (E.S.); (N.A.); (P.O.); (D.P.)
| | - Daniil M. Bobrovskiy
- Faculty of Bioengineering and Bioinformatics, Moscow State University, 119234 Moscow, Russia;
| | - Nickolay Anikanov
- V. Zelman Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia; (A.T.); (E.S.); (N.A.); (P.O.); (D.P.)
| | - Polina Ogurtsova
- V. Zelman Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia; (A.T.); (E.S.); (N.A.); (P.O.); (D.P.)
| | - Dong Ik Park
- Proteomics and Biomarkers, Max Planck Institute of Psychiatry, 80804 Munich, Germany;
| | - Anja K. E. Horn
- Institute of Anatomy and Cell Biology, Ludwig-Maximilians University, 80336 Munich, Germany;
| | - Daria Petrova
- V. Zelman Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia; (A.T.); (E.S.); (N.A.); (P.O.); (D.P.)
| | - Ekaterina Khrameeva
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia
- Correspondence: (E.K.); (M.S.G.); (C.W.T.); (P.K.)
| | - Mari S. Golub
- California National Primate Research Center, University of California, Davis, CA 95616, USA
- Correspondence: (E.K.); (M.S.G.); (C.W.T.); (P.K.)
| | - Christoph W. Turck
- Proteomics and Biomarkers, Max Planck Institute of Psychiatry, 80804 Munich, Germany;
- Correspondence: (E.K.); (M.S.G.); (C.W.T.); (P.K.)
| | - Philipp Khaitovich
- V. Zelman Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia; (A.T.); (E.S.); (N.A.); (P.O.); (D.P.)
- Correspondence: (E.K.); (M.S.G.); (C.W.T.); (P.K.)
| |
Collapse
|
31
|
Yu H, Qin X, Yu Z, Chen Y, Tang L, Shan W. Effects of high-fat diet on the formation of depressive-like behavior in mice. Food Funct 2021; 12:6416-6431. [PMID: 34076000 DOI: 10.1039/d1fo00044f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Depression is an important global health issue that is associated with serious physical and mental health consequences. The field of nutritional psychiatry has generated observational and efficacy data supporting a role for healthy dietary patterns in depression. Here, we aim to evaluate the effects of high-fat diet (HFD) consumption on depressive-like behaviors. BALB/c mice were grouped randomly: control, chronic restraint stress (CRS), HFD and CRS + HFD groups. The depressive-like behavior was evaluated using behavioral tests. The serotonin content in murine brain tissue and blood lipid concentrations were detected by ELISA. The fatty acid content in the liver, adipose tissue of epididymis, brain tissue, and serum of mice was determined by gas chromatography (GC). Expression of the fatty acid synthesis pathway-related enzymes at the mRNA level was analyzed by qRT-PCR. The results indicated that a high-fat diet could promote depressive-like behavior. In comparison with regular feeding, concentrations of blood lipids were significantly changed in the HFD group. Correlation analysis implied that high-density lipoprotein cholesterol (HDL-c) and low-density lipoprotein cholesterol (LDL-c) were closely related to depressive-like behavior. Based on fatty acid analysis, the palmitoleic acid, linoleic acid, oleic acid, and arachidonic acid content was remarkably changed in mice with depressive-like behavior. In addition, acetyl-CoA carboxylase (ACC), stearoyl-CoA desaturase-1 (SCD1), fatty acid desaturase 1 (FADS1), and fatty acid desaturase 2 (FADS2) expression, which are involved in de novo fatty acid synthesis, desaturation of fatty acids, and arachidonic acid synthesis, were strengthened in HFD mice with depressive-like behavior. Therefore, we postulated that the disorder of lipid metabolism induced by HFD consumption accelerated the development of depressive-like behavior.
Collapse
Affiliation(s)
- Haining Yu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China.
| | | | | | | | | | | |
Collapse
|
32
|
Hu Y, Chen C, Wang Y, Yang W, Wang Y, Zhu W, Yan C, Liu P. The effects of KaiXinSan on depression and its association with lipid profiles: A randomized, double-blinded, placebo-controlled trial. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 83:153467. [PMID: 33516143 DOI: 10.1016/j.phymed.2021.153467] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Traditional Chinese medicine (TCM) KaiXinSan (KXS) has been used to treat depressed patients for a long time, but its potential underlying mechanisms have not been fully understood. HYPOTHESIS KXS could mitigate symptoms of patients with atypical depression at least partly via regulating lipid equilibrium. METHODS Patients meeting DSM-IV criteria for mild or moderate depression were assigned into placebo (N = 68) or KXS 3.2 g/day (N = 66) groups in a randomized, double-blinded, placebo-controlled, parallel clinical trial to investigate the anti-depressive efficacy of KXS and its association with serum lipid profile. RESULTS The HAMD score and SDS score at 8 weeks were significantly improved in KXS-treated patients the N-BACK accuracy rate was also increased after 8 weeks of KXS treatment compared with baseline. These results indicated that KXS not only improved the specific symptoms of depression, but also had a beneficial effect on cognitive function related working memory. More importantly, KXS treatment improved patients' lipid profile by reducing the ratios of LDL/HDL and ApoB/ApoA1 (p < 0.05), as well as ApoC3 level. Moreover, subgroup analysis found that HAMD score was significantly higher in patients with high lipid profile than in those with normal lipid profile, and lipid improvement after 8 weeks of KXS treatment was more obvious in depressed patients with high lipid profile than with normal lipid profile. CONCLUSION KXS could mitigate symptoms of patients with minor and modest depression at least partly via regulating lipid equilibrium. Its might shed light that KXS may likely contributes to depressed patients with other cardio-metabolic diseases.
Collapse
Affiliation(s)
- Yuan Hu
- Department of Pharmacy, Medical Supplier Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Chao Chen
- Department of Pharmacy, Medical Supplier Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Yichen Wang
- Department of Pharmacy, Medical Supplier Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Wenshan Yang
- Department of Pharmacy, Medical Supplier Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Yuanbo Wang
- Department of Pharmacy, Medical Supplier Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Weiyu Zhu
- Department of Pharmacy, Medical Supplier Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Can Yan
- Department of Basic Theory of TCM, College of Basic Medicine Sciences, Guangzhou University of Chinese Medicine.
| | - Ping Liu
- Department of Pharmacy, Medical Supplier Center, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
33
|
Xin J, Yan S, Hong X, Zhang H, Zha J. Environmentally relevant concentrations of carbamazepine induced lipid metabolism disorder of Chinese rare minnow (Gobiocypris rarus) in a gender-specific pattern. CHEMOSPHERE 2021; 265:129080. [PMID: 33261836 DOI: 10.1016/j.chemosphere.2020.129080] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/01/2020] [Accepted: 11/19/2020] [Indexed: 06/12/2023]
Abstract
Carbamazepine (CBZ), an anticonvulsant and mood stabilizer, is ubiquitous distributed in aquatic environment. Though the toxicity and endocrine disrupting effect of CBZ on non-target organisms have been studied, its lipotoxity are scarcely known. To assess the lipotoxicity of CBZ, 2-month-old Chinese rare minnow were exposed to 0, 1, 10, and 100 μg/L CBZ for 90 d. Obvious dyslipidemia was observed after 30 d and 90 d exposure, whereas overt hyperlipidemia was observed in males at 100 μg/L treatments. Severe lipid droplet accumulation in livers was observed at 10 and 100 μg/L treatments for 30 d and in females, whereas those was observed at all treatments in males. In addition, serious mitochondria damage was observed in males at 100 μg/L treatments. After 90 d exposure, the enzyme activities of FAS and ACCα were significantly increased at 10 and 100 μg/L treatments, whereas HMGCR were markedly increased at 100 μg/L treatments (p < 0.05). However, ACCβ were markedly decreased in females at 10 and 100 μg/L treatments and in males at all treatments (p < 0.05). The transcription levels of fasn, accα, hmgcrα, fdft1, idi1, plin1, plin2, caveolin1, and caveolin2 were significantly increased at 100 μg/L treatments (p < 0.05). Moreover, the body weight was obviously increased at 10 and 100 μg/L treatments in males (p < 0.05). Our results confirmed that environmental relevant concentrations CBZ induced lipid metabolism disorder and mitochondria damage of Chinese rare minnow in a gender-specific pattern, which provided a new insight into the lipotoxicity mechanism of CBZ.
Collapse
Affiliation(s)
- Jiajing Xin
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; Beijing Key Laboratory of Industrial Wastewater Treatment and Reuse, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100085, China
| | - Saihong Yan
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; Beijing Key Laboratory of Industrial Wastewater Treatment and Reuse, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100085, China
| | - Xiangsheng Hong
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; Beijing Key Laboratory of Industrial Wastewater Treatment and Reuse, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100085, China
| | - Huan Zhang
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, College of Fisheries, Huazhong Agriculture University, Wuhan, 430070, China
| | - Jinmiao Zha
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; Beijing Key Laboratory of Industrial Wastewater Treatment and Reuse, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100085, China.
| |
Collapse
|
34
|
Ayyash A, Holloway AC. Fluoxetine-induced hepatic lipid accumulation is linked to elevated serotonin production. Can J Physiol Pharmacol 2021; 99:983-988. [PMID: 33517848 DOI: 10.1139/cjpp-2020-0721] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Fluoxetine, a commonly prescribed selective serotonin reuptake inhibitor antidepressant, has been shown to increase hepatic lipid accumulation, a key factor in the development of nonalcoholic fatty liver disease. Interestingly, fluoxetine has also been reported to increase peripheral serotonin synthesis. As emerging evidence suggests that serotonin may be involved in the development of nonalcoholic fatty liver disease, we sought to determine if fluoxetine-induced hepatic lipid accumulation is mediated via altered serotonin production. Fluoxetine treatment increased lipid accumulation in association with increased mRNA expression of tryptophan hydroxylase 1 (Tph1, serotonin biosynthetic enzyme) and intracellular serotonin content. Serotonin alone had a similar effect to increase lipid accumulation. Moreover, blocking serotonin synthesis reversed the fluoxetine-induced increases in lipid accumulation. Collectively, these data suggest that fluoxetine-induced lipid accumulation can be mediated, in part, by elevated serotonin production. These results suggest a potential therapeutic target to ameliorate the adverse metabolic effects of fluoxetine exposure.
Collapse
Affiliation(s)
- Ahmed Ayyash
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON L8S 4K1, Canada.,Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Alison C Holloway
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON L8S 4K1, Canada.,Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
35
|
Todorović Vukotić N, Đorđević J, Pejić S, Đorđević N, Pajović SB. Antidepressants- and antipsychotics-induced hepatotoxicity. Arch Toxicol 2021; 95:767-789. [PMID: 33398419 PMCID: PMC7781826 DOI: 10.1007/s00204-020-02963-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023]
Abstract
Drug-induced liver injury (DILI) is a serious health burden. It has diverse clinical presentations that can escalate to acute liver failure. The worldwide increase in the use of psychotropic drugs, their long-term use on a daily basis, common comorbidities of psychiatric and metabolic disorders, and polypharmacy in psychiatric patients increase the incidence of psychotropics-induced DILI. During the last 2 decades, hepatotoxicity of various antidepressants (ADs) and antipsychotics (APs) received much attention. Comprehensive review and discussion of accumulated literature data concerning this issue are performed in this study, as hepatotoxic effects of most commonly prescribed ADs and APs are classified, described, and discussed. The review focuses on ADs and APs characterized by the risk of causing liver damage and highlights the ones found to cause life-threatening or severe DILI cases. In parallel, an overview of hepatic oxidative stress, inflammation, and steatosis underlying DILI is provided, followed by extensive review and discussion of the pathophysiology of AD- and AP-induced DILI revealed in case reports, and animal and in vitro studies. The consequences of some ADs and APs ability to affect drug-metabolizing enzymes and therefore provoke drug–drug interactions are also addressed. Continuous collecting of data on drugs, mechanisms, and risk factors for DILI, as well as critical data reviewing, is crucial for easier DILI diagnosis and more efficient risk assessment of AD- and AP-induced DILI. Higher awareness of ADs and APs hepatotoxicity is the prerequisite for their safe use and optimal dosing.
Collapse
Affiliation(s)
- Nevena Todorović Vukotić
- Department of Molecular Biology and Endocrinology, "Vinča" Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 12-14 Mike Petrovića Alasa, P.O. Box 522-090, 11000, Belgrade, Serbia.
| | - Jelena Đorđević
- Institute of Physiology and Biochemistry "Ivan Đaja", Faculty of Biology, University of Belgrade, 16 Studentski Trg, 11000, Belgrade, Serbia
| | - Snežana Pejić
- Department of Molecular Biology and Endocrinology, "Vinča" Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 12-14 Mike Petrovića Alasa, P.O. Box 522-090, 11000, Belgrade, Serbia
| | - Neda Đorđević
- Department of Molecular Biology and Endocrinology, "Vinča" Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 12-14 Mike Petrovića Alasa, P.O. Box 522-090, 11000, Belgrade, Serbia
| | - Snežana B Pajović
- Department of Molecular Biology and Endocrinology, "Vinča" Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 12-14 Mike Petrovića Alasa, P.O. Box 522-090, 11000, Belgrade, Serbia.,Faculty of Medicine, University of Niš, 81 Blvd. Dr. Zorana Đinđića, 18000, Niš, Serbia
| |
Collapse
|
36
|
Lu S, Ma Z, Gu Y, Li P, Chen Y, Bai M, Zhou H, Yang X, Jiang H. Downregulation of glucose‐6‐phosphatase expression contributes to fluoxetine‐induced hepatic steatosis. J Appl Toxicol 2020; 41:1232-1240. [PMID: 33179799 DOI: 10.1002/jat.4109] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/21/2020] [Accepted: 10/26/2020] [Indexed: 12/21/2022]
Affiliation(s)
- Shuanghui Lu
- Institute of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences Zhejiang University Hangzhou China
| | - Zhiyuan Ma
- Institute of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences Zhejiang University Hangzhou China
- Affiliated Hangzhou First People's Hospital, College of Medicine Zhejiang University Hangzhou China
| | - Yong Gu
- Institute of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences Zhejiang University Hangzhou China
| | - Ping Li
- Institute of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences Zhejiang University Hangzhou China
| | - Yingchun Chen
- Institute of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences Zhejiang University Hangzhou China
| | - Mengru Bai
- Institute of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences Zhejiang University Hangzhou China
- Affiliated Hangzhou First People's Hospital, College of Medicine Zhejiang University Hangzhou China
| | - Hui Zhou
- Institute of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences Zhejiang University Hangzhou China
| | - Xi Yang
- The First Affiliated Hospital, College of Medicine Zhejiang University Hangzhou China
| | - Huidi Jiang
- Institute of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences Zhejiang University Hangzhou China
| |
Collapse
|
37
|
Yang H, Cao Q, Xiong X, Zhao P, Shen D, Zhang Y, Zhang N. Fluoxetine regulates glucose and lipid metabolism via the PI3K‑AKT signaling pathway in diabetic rats. Mol Med Rep 2020; 22:3073-3080. [PMID: 32945450 PMCID: PMC7453494 DOI: 10.3892/mmr.2020.11416] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 11/01/2018] [Indexed: 12/02/2022] Open
Abstract
Diabetes mellitus poses a major threat towards global heath due to a lack of effective treatment. Fluoxetine hydrochloride, a selective 5-hydroxytryptamine reuptake inhibitor, is the most commonly used antidepressant in clinical therapy; however, the potential molecular mechanisms of fluoxetine in diabetes remain unknown. In the present study, reduced glucose, total cholesterol and triglyceride levels and lipid metabolism, as well as upregulated proliferator-activated receptor γ, fatty acid synthase and lipoprotein lipase, and downregulated sterol regulatory element-binding protein 1-c were detected in rats with streptozotocin (STZ)-induced diabetes following treatment with fluoxetine. Furthermore, fluoxetine significantly inhibited the expression levels of glucose metabolism-associated proteins in liver tissues, including glycogen synthase kinase 3β (GSK-3β), glucose-6 phosphatase catalytic subunit (G6PC), phosphoenolpyruvate carboxykinase (PEPCK) and forkhead box protein O1 (FOXO1). In addition, fluoxetine treatment notably attenuated morphological liver damage in rats with STZ-induced diabetes. Additionally, fluoxetine could inhibit the phosphatidylinositol 3-kinase-protein kinase B (PI3K-AKT) signaling pathway, whereas LY294002, a specific inhibitor of PI3K, suppressed the function of PI3K-AKT signaling and suppressed the expression levels of glucose metabolism-associated proteins, including GSK-3β, G6PC, PEPCK and FOXO1 in BRL-3A cells. The results of the present study revealed that fluoxetine may regulate glucose and lipid metabolism via the PI3K-AKT signaling pathway in diabetic rats.
Collapse
Affiliation(s)
- Hailong Yang
- Department of Clinical Psychology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Qiuyun Cao
- Department of Clinical Psychology, Nanjing Drum Tower Hospital Affiliated to Nanjing University Medical College, Nanjing, Jiangsu 210000, P.R. China
| | - Xiaolu Xiong
- Department of Endocrinology, Nanjing Drum Tower Hospital Affiliated to Nanjing University Medical College, Nanjing, Jiangsu 210000, P.R. China
| | - Peng Zhao
- Department of Clinical Psychology, Nanjing Drum Tower Hospital Affiliated to Nanjing University Medical College, Nanjing, Jiangsu 210000, P.R. China
| | - Diwen Shen
- Department of Clinical Psychology, Nanjing Drum Tower Hospital Affiliated to Nanjing University Medical College, Nanjing, Jiangsu 210000, P.R. China
| | - Yuzhe Zhang
- Department of Clinical Psychology, Nanjing Drum Tower Hospital Affiliated to Nanjing University Medical College, Nanjing, Jiangsu 210000, P.R. China
| | - Ning Zhang
- Department of Clinical Psychology, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| |
Collapse
|
38
|
Feijão E, Cruz de Carvalho R, Duarte IA, Matos AR, Cabrita MT, Novais SC, Lemos MFL, Caçador I, Marques JC, Reis-Santos P, Fonseca VF, Duarte B. Fluoxetine Arrests Growth of the Model Diatom Phaeodactylum tricornutum by Increasing Oxidative Stress and Altering Energetic and Lipid Metabolism. Front Microbiol 2020; 11:1803. [PMID: 32849412 PMCID: PMC7411086 DOI: 10.3389/fmicb.2020.01803] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 07/09/2020] [Indexed: 11/13/2022] Open
Abstract
Pharmaceutical residues impose a new and emerging threat to aquatic environments and its biota. One of the most commonly prescribed pharmaceuticals is the antidepressant fluoxetine, a selective serotonin re-uptake inhibitor that has been frequently detected, in concentrations up to 40 μg L–1, in aquatic ecosystems. The present study aims to investigate the ecotoxicity of fluoxetine at environmentally relevant concentrations (0.3, 0.6, 20, 40, and 80 μg L–1) on cell energy and lipid metabolism, as well as oxidative stress biomarkers in the model diatom Phaeodactylum tricornutum. Exposure to higher concentrations of fluoxetine negatively affected cell density and photosynthesis through a decrease in the active PSII reaction centers. Stress response mechanisms, like β-carotene (β-car) production and antioxidant enzymes [superoxide dismutase (SOD) and ascorbate peroxidase (APX)] up-regulation were triggered, likely as a positive feedback mechanism toward formation of fluoxetine-induced reactive oxygen species. Lipid peroxidation products increased greatly at the highest fluoxetine concentration whereas no variation in the relative amounts of long chain polyunsaturated fatty acids (LC-PUFAs) was observed. However, monogalactosyldiacylglycerol-characteristic fatty acids such as C16:2 and C16:3 increased, suggesting an interaction between light harvesting pigments, lipid environment, and photosynthesis stabilization. Using a canonical multivariate analysis, it was possible to evaluate the efficiency of the application of bio-optical and biochemical techniques as potential fluoxetine exposure biomarkers in P. tricornutum. An overall classification efficiency to the different levels of fluoxetine exposure of 61.1 and 88.9% were obtained for bio-optical and fatty acids profiles, respectively, with different resolution degrees highlighting these parameters as potential efficient biomarkers. Additionally, the negative impact of this pharmaceutical molecule on the primary productivity is also evident alongside with an increase in respiratory oxygen consumption. From the ecological point of view, reduction in diatom biomass due to continued exposure to fluoxetine may severely impact estuarine and coastal trophic webs, by both a reduction in oxygen primary productivity and reduced availability of key fatty acids to the dependent heterotrophic upper levels.
Collapse
Affiliation(s)
- Eduardo Feijão
- MARE - Marine and Environmental Sciences Centre, Faculdade de Ciências da Universidade de Lisboa, Lisbon, Portugal
| | - Ricardo Cruz de Carvalho
- MARE - Marine and Environmental Sciences Centre, Faculdade de Ciências da Universidade de Lisboa, Lisbon, Portugal.,cE3c - Centre for Ecology, Evolution and Environmental Changes, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Irina A Duarte
- MARE - Marine and Environmental Sciences Centre, Faculdade de Ciências da Universidade de Lisboa, Lisbon, Portugal
| | - Ana Rita Matos
- BioISI - Biosystems and Integrative Sciences Institute, Plant Functional Genomics Group, Departamento de Biologia Vegetal, Faculdade de Ciências da Universidade de Lisboa, Lisbon, Portugal
| | - Maria Teresa Cabrita
- Centro de Estudos Geográficos, Instituto de Geografia e Ordenamento do Território, University of Lisbon, Lisbon, Portugal
| | - Sara C Novais
- MARE - Marine and Environmental Sciences Centre, ESTM, Politécnico de Leiria, Peniche, Portugal
| | - Marco F L Lemos
- MARE - Marine and Environmental Sciences Centre, ESTM, Politécnico de Leiria, Peniche, Portugal
| | - Isabel Caçador
- MARE - Marine and Environmental Sciences Centre, Faculdade de Ciências da Universidade de Lisboa, Lisbon, Portugal
| | - João Carlos Marques
- MARE - Marine and Environmental Sciences Centre, Department of Zoology, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Patrick Reis-Santos
- MARE - Marine and Environmental Sciences Centre, Faculdade de Ciências da Universidade de Lisboa, Lisbon, Portugal.,Southern Seas Ecology Laboratories, School of Biological Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Vanessa F Fonseca
- MARE - Marine and Environmental Sciences Centre, Faculdade de Ciências da Universidade de Lisboa, Lisbon, Portugal.,Departamento de Biologia Animal, Faculdade de Ciências da Universidade de Lisboa, Lisbon, Portugal
| | - Bernardo Duarte
- MARE - Marine and Environmental Sciences Centre, Faculdade de Ciências da Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
39
|
Fittipaldi EODS, Andrade ADD, Santos ACO, Campos S, Fernandes J, Catanho MTJDA. Depressive Symptoms are Associated with High Levels of Serum Low-Density Lipoprotein Cholesterol in Older Adults with Type 2 Diabetes Mellitus. Arq Bras Cardiol 2020; 115:462-467. [PMID: 32696856 PMCID: PMC9363090 DOI: 10.36660/abc.20190404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 08/18/2019] [Indexed: 11/18/2022] Open
Abstract
Fundamento O Diabetes Mellitus Tipo 2 (DMT2) é comum nos idosos, que também apresentam um nível elevado de fatores de risco para doenças cardiovasculares (DCVs), tais como dislipidemia. Entretanto, o papel da depressão nos pacientes com DMT2 e sua relação com fatores de risco para DCV são pouco estudados. Objetivo O objetivo do presente estudo foi investigar a relação entre sintomas depressivos (SDs) e fatores de risco cardiovascular conhecidos em idosos comunitários portadores de DMT2. Métodos Trata-se de um estudo transversal, no qual foram incluídos 85 idosos comunitários com DMT2. Os SDs foram avaliados através da Escala de Depressão Geriátrica de Yesavage, em versão reduzida (GDS-15). Os seguintes fatores de risco cardiovascular foram avaliados: pressão arterial sistólica (PAS) e diastólica (PAD), glicose plasmática em jejum (GPJ), perfil lipídico (triglicerídeos séricos (TG), colesterol total sérico (CT), colesterol sérico de lipoproteína de baixa densidade (LDL-C) e colesterol sérico de lipoproteína de baixa densidade (HDL-C)) e índice de massa corporal (IMC). A análise de regressão múltipla de Poisson foi utilizada para avaliar a associação entre os SDs e cada fator de risco cardiovascular ajustado por sexo, idade, tempo em atividades físicas moderadas e status funcional. O nível de significância adotado para a análise foi de 5%. Resultados Dentre todos os fatores de risco analisados, apenas o aumento de LDL-C apresentou uma correlação com níveis elevados de SD (RP=1,005; IC95% 1,002-1,008). Foi observada uma associação significativa entre os níveis de HDL-C (RP=0,99; IC95% 0,98-0,99) e a PAS (RP=1,009; IC95% 1,004-1,014). Conclusão Nos idosos com DMT2, a presença de SD foi associada a níveis de LDL-C, HDL-C e PAS, mesmo após o ajuste por sexo, idade, nível de atividade física e capacidade funcional. (Arq Bras Cardiol. 2020; 115(3):462-467)
Collapse
|
40
|
Metformin ameliorates stress-induced depression-like behaviors via enhancing the expression of BDNF by activating AMPK/CREB-mediated histone acetylation. J Affect Disord 2020; 260:302-313. [PMID: 31521867 DOI: 10.1016/j.jad.2019.09.013] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 07/22/2019] [Accepted: 09/02/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Metformin, a first-line antiglycemic drug, has been reported to have anti-depressant effects in patients with type 2 diabetes; however, its exact role and underlying mechanism still need to be investigated. METHOD C57BL/6J mice were subjected to the Chronic social defeat stress (SDS) and drug administration (Control + Vehicle, SDS + Vehicle, SDS + MET (200 mg kg-1), SDS + FLUOX (3 mg kg-1), SDS + MET + FLUOX). And the depression phenotypes were evaluated by the sucrose preference test, social interaction, tail suspension test and forced swimming test. The potential mechanisms underlying the effects of metformin on depression was discussed by using Chromatin immunoprecipitation, Quantitative real-time PCR mRNA expression analysis and Western blot in vivo and in primary cultured hippocampal neurons. RESULT The metformin treatment counteracted the development of depression-like behaviors in mice suffering SDS when administered alone and enhanced the anti-depressant effect of fluoxetine when combined with fluoxetine. Further RNA sequencing analysis revealed that metformin treatment prevented the transcriptional changes in the medial prefrontal cortex (mPFC) of the animals and Golgi staining indicated favorable morphological changes in the neurite plasticity of CA1 pyramidal neurons, which approximated to those found in unstressed mice. At a molecular level, metformin significantly upregulated the expression of the brain-derived neurotrophic factor (BDNF) by increasing the histone acetylation along with the BDNF promoter, which was attributed to the activation of AMP-activated protein kinase (AMPK) and cAMP-response element binding protein (CREB). CONCLUSION Our findings suggest that metformin can produce antidepressant effects, which provides empirical insights into the clinical value of metformin in the prevention and therapy of depression.
Collapse
|
41
|
Yao T, Cui Q, Liu Z, Wang C, Zhang Q, Wang G. Metabolomic evidence for the therapeutic effect of gentiopicroside in a corticosterone-induced model of depression. Biomed Pharmacother 2019; 120:109549. [PMID: 31655313 DOI: 10.1016/j.biopha.2019.109549] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/07/2019] [Accepted: 10/08/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Depression is a disease that seriously threatens the quality of human life. To explore the effect of gentiopicroside on depression, this study investigated the therapeutic effect of gentiopicroside on corticosterone-induced depressionin vivo and in vitro by using metabolomic methods. METHODS A total of 36 rats were randomly assigned to three groups: a normal group, model group (depression), and treatment group (depression + gentiopicroside). Corticosterone was administrated to induce depression-like model rats. Morris water maze test was used to validated the behavior performance. The hippocampus of rats was obtained for metabolomic detection. Metabolites that were differentially expressed between the groups were extracted for Heatmap, Go, and pathway enrichment analyses. Finally, neuronal cells were cultured and examined to validated the effect of gentiopicroside. RESULTS Corticosterone injured rats learning capacity, and decreased the levels of 5-HT, and reversed by gentiopicroside delivery. Metabolites obtained from the hippocampus of rats in the three groups were subjected to a principal component analysis (PCA). Go and pathway enrichment analyses revealed the involvement of sphingolipid metabolism et al. Gentiopicroside could inhibit apoptosis caused by corticosterone, and also decrease neuronal cell proliferation and BDNF levels in vitro. Arachidonic acid (ARA) reversed the protective effect of gentiopicroside on neuronal cells. CONCLUSION These findings suggest that gentiopicroside reduces apoptosis and increases the proliferation of hippocampus cells in depressed animals by regulating metabolites. Moreover, our study provides a new basis for the clinical treatment of depression and demonstrates the potential efficacy of gentiopicroside in this area of pathology.
Collapse
Affiliation(s)
- Tao Yao
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qin Cui
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhichao Liu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Cuifang Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qi Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
42
|
Zhou YF, Feng L, Liu XM, Tao X, Wang LS, Zhang MD, Wang Z, Chen SG, Chang Q. Urinary metabolic disturbance in the olfactory bulbectomized rats and the modulatory effects of fluoxetine. Life Sci 2019; 234:116751. [DOI: 10.1016/j.lfs.2019.116751] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/02/2019] [Accepted: 08/10/2019] [Indexed: 12/11/2022]
|
43
|
Zhang K, Wang X, Tu J, Rong H, Werz O, Chen X. The interplay between depression and tuberculosis. J Leukoc Biol 2019; 106:749-757. [PMID: 31254317 DOI: 10.1002/jlb.mr0119-023r] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/19/2019] [Accepted: 06/07/2019] [Indexed: 02/06/2023] Open
Abstract
Depression is a major mental health condition and is expected be the most debilitating and widespread health disorder by 2030. Tuberculosis (TB) is also a leading cause of morbidity and mortality worldwide and interestingly, is a common comorbidity of depression. As such, much attention has been paid to the association between these 2 pathologies. Based on clinical reports, the association between TB and depression seems to be bidirectional, with a substantial overlap in symptoms between the 2 conditions. TB infection or reactivation may precipitate depression, likely as a consequence of the host's inflammatory response and/or dysregulation of the hypothalamic-pituitary-adrenal axis. Nevertheless, few studies have considered whether patients with depression are at a higher risk for TB. In this review, we discuss the hypotheses on the association between depression and TB, highlighting the immuno-inflammatory response and lipid metabolism as potential mechanisms. Improving our understanding of the interplay between these 2 disorders should help guide TB clinical care and prevention both in patients with comorbid depression and in the general population.
Collapse
Affiliation(s)
- Kehong Zhang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China.,Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| | - Xin Wang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Jie Tu
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institutes of Advanced Technology, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Chinese Academy of Sciences, Shenzhen, China
| | - Han Rong
- Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, China
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| | - Xinchun Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| |
Collapse
|
44
|
Hu MZ, Wang AR, Zhao ZY, Chen XY, Li YB, Liu B. Antidepressant-like effects of paeoniflorin on post-stroke depression in a rat model. Neurol Res 2019; 41:446-455. [PMID: 30759063 DOI: 10.1080/01616412.2019.1576361] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 01/26/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Post-stroke depression (PSD) is one of the most prevalent emotional disorders after stroke and often results in poor outcomes. However, the underlying physiopathologic mechanism and effective treatment of PSD remain poorly elucidated. OBJECTIVE To investigate whether paeoniflorin has antidepressant-like activity in a rat model of PSD. METHODS Rats were randomly divided into four groups: sham-operated control (Sham), PSD, paeoniflorin (with PSD) and fluoxetine group(with PSD). PSD was developed by the right middle cerebral artery occlusion followed 21 days chronic unpredictable mild stress combined (CUMS) with raised alone. Tests of sucrose preference and open field were used to assess the depression-like behavior. Neurological function was evaluated by neurological deficit score and beam balance test. Expression of phosphorylated CREB (p-CREB) and brain-derived neurotrophic factor (BDNF) in the CA1 region of the hippocampal complex was evaluated by western blot and immunofluorescence. RESULTS Te depressive-like behaviors markedly improved after paeoniflorin and fluoxetine treatment. Furthermore, paeoniflorin treatment significantly increased BDNF and p-CREB expression in the CA1 region. CONCLUSIONS Observed results suggested that paeoniflorin could ameliorate the symptoms and improve the functional capability of PSD rats, similar to the effect of fluoxetine. ABBREVIATIONS PSD: post-stroke depression; CUMS: chronic unpredictable mild stress stimulation; MCAO: middle cerebral artery occlusion; OFT: open field test; SPT: sucrose preference test, NDS: neurological deficit score, BBT: beam balance test; BDNF: brain-derived neurotrophic factor protein; p-CREB: phosphorylated Cyclic-AMP responsive element binding protein.
Collapse
Affiliation(s)
- Ming-Zhe Hu
- a The Second Clinical College of Guangzhou University of Chinese Medicine , Guangzhou , China
| | - An-Rong Wang
- b Shandong University of Traditional Chinese Medicine , Jinan , China
| | - Ze-Yu Zhao
- c School of Clinical Medicine , Weifang Medical University , Weifang , China
| | - Xiang-Yan Chen
- d Department of Internal Medicine , Zibo City Hospital of Traditional Chinese Medicine , Zibo , China
| | - Yan-Bin Li
- e Department of Neurology, Shandong Provincial Qianfoshan Hospital , Shandong University , Jinan , China
| | - Bin Liu
- e Department of Neurology, Shandong Provincial Qianfoshan Hospital , Shandong University , Jinan , China
| |
Collapse
|
45
|
Chen XX, Xu YY, Wu R, Chen Z, Fang K, Han YX, Yu Y, Huang LL, Peng L, Ge JF. Resveratrol Reduces Glucolipid Metabolic Dysfunction and Learning and Memory Impairment in a NAFLD Rat Model: Involvement in Regulating the Imbalance of Nesfatin-1 Abundance and Copine 6 Expression. Front Endocrinol (Lausanne) 2019; 10:434. [PMID: 31338065 PMCID: PMC6629830 DOI: 10.3389/fendo.2019.00434] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 06/17/2019] [Indexed: 12/12/2022] Open
Abstract
Resveratrol (RES) is a polyphenolic compound, and our previous results have demonstrated its neuroprotective effect in a series of animal models. The aim of this study was to investigate its potential effect on a nonalcoholic fatty liver disease (NAFLD) rat model. The parameters of liver function and glucose and lipid metabolism were measured. Behavior performance was observed via the open field test (OFT), the sucrose preference test (SPT), the elevated plus maze (EPM), the forced swimming test (FST), and the Morris water maze (MWM). The protein expression levels of Copine 6, p-catenin, catenin, p-glycogen synthase kinase-3beta (GSK3β), GSK3β, and cyclin D1 in the hippocampus and prefrontal cortex (PFC) were detected using Western blotting. The results showed that RES could reverse nesfatin-1-related impairment of liver function and glucolipid metabolism, as indicated by the decreased plasma concentrations of alanine aminotransferase (ALT), aspartate aminotransferase (AST), total bilirubin (TBIL), direct bilirubin (DBIL), indirect bilirubin (IBIL), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), glucose, insulin, and nesfatin-1; increase the plasma level of high-density lipoprotein cholesterol (HDL-C); and reduce hepatocyte steatosis in NAFLD rats. Although there was no significant difference among groups with regard to performance in the OFT, EPM, and FST tasks, RES-treated NAFLD rats showed an increased sucrose preference index in the SPT and improved learning and memory ability in the MWM task. Furthermore, the imbalanced protein expression levels of Copine 6, p-catenin, and p-GSK3β in the hippocampus and PFC of NAFLD rats were also restored to normal by treatment with RES. These results suggested that four consecutive weeks of RES treatment not only ameliorated glucolipid metabolic impairment and liver dysfunction in the NAFLD rat model but also mitigated the attendant behavioral and cognitive impairments. In addition to the mediating role of nesfatin-1, the mechanism underlying the therapeutic effect of RES on NAFLD might be associated with its ability to regulate the imbalanced expression level of Copine 6 and the Wnt signaling pathway in the hippocampus and PFC.
Collapse
Affiliation(s)
- Xing-Xing Chen
- School of Pharmacy, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Ya-Yun Xu
- School of Pharmacy, Anhui Medical University, Hefei, China
- Department of Pharmacy, The Fourth People's Hospital in Hefei, Hefei, China
| | - Rui Wu
- School of Pharmacy, Anhui Medical University, Hefei, China
- Department of Pharmacy, The People's Hospital of Huangshan, Huangshan, China
| | - Zheng Chen
- School of Pharmacy, Anhui Medical University, Hefei, China
- Department of Pharmacy, Lujiang County Hospital of Traditional Chinese Medicine, Hefei, China
| | - Ke Fang
- School of Pharmacy, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Yin-Xiu Han
- School of Pharmacy, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Yue Yu
- School of Pharmacy, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Ling-Ling Huang
- School of Pharmacy, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Lei Peng
- School of Pharmacy, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Jin-Fang Ge
- School of Pharmacy, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
- *Correspondence: Jin-Fang Ge
| |
Collapse
|
46
|
Zhou H, Tang J, Zhang J, Chen B, Kan J, Zhang W, Zhou J, Ma H. A red lysosome-targeted fluorescent probe for carboxylesterase detection and bioimaging. J Mater Chem B 2019. [DOI: 10.1039/c9tb00310j] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A red lysosome-targeting probe for carboxylesterase activity has been successfully applied in complex biological samples.
Collapse
Affiliation(s)
- Hui Zhou
- College of Pharmacy
- Weifang Medical University
- Weifang
- China
| | - Jinbao Tang
- College of Pharmacy
- Weifang Medical University
- Weifang
- China
| | - Jie Zhang
- College of Pharmacy
- Weifang Medical University
- Weifang
- China
| | - Bochao Chen
- College of Pharmacy
- Weifang Medical University
- Weifang
- China
| | - Jianfei Kan
- College of Pharmacy
- Weifang Medical University
- Weifang
- China
| | - Weifen Zhang
- College of Pharmacy
- Weifang Medical University
- Weifang
- China
| | - Jin Zhou
- College of Pharmacy
- Weifang Medical University
- Weifang
- China
| | - Huimin Ma
- Beijing National laboratory for Molecular Sciences
- Key Laboratory of Analytical Chemistry for Living Biosystems
- Institute of Chemistry
- Chinese Academy of Sciences
- Bejing 100190
| |
Collapse
|