1
|
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
|
2
|
Peña JE, Corbett BF, Tamminga CA, Bhatnagar S, Hitti FL. Investigating Resistance to Antidepressants in Animal Models. Neuroscience 2024; 548:69-80. [PMID: 38697464 DOI: 10.1016/j.neuroscience.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/12/2024] [Accepted: 04/25/2024] [Indexed: 05/05/2024]
Abstract
Major depressive disorder is one of the most prevalent psychiatric diseases, and up to 30-40% of patients remain symptomatic despite treatment. Novel therapies are sorely needed, and animal models may be used to elucidate fundamental neurobiological processes that contribute to human disease states. We conducted a systematic review of current preclinical approaches to investigating treatment resistance with the goal of describing a path forward for improving our understanding of treatment resistant depression. We conducted a broad literature search to identify studies relevant to the preclinical investigation of treatment resistant depression. We followed PRISMA (Preferred Reporting Items for Systemic Reviews and Meta-Analyses) guidelines and included all relevant studies. We identified 467 studies in our initial search. Of these studies, we included 69 in our systematic review after applying our inclusion/exclusion criteria. We identified 10 broad strategies for investigating treatment resistance in animal models. Stress hormone administration was the most commonly used model, and the most common behavioral test was the forced swim test. We systematically identified and reviewed current approaches for gaining insight into the neurobiology underlying treatment resistant depression using animal models. Each approach has its advantages and disadvantages, but all require careful consideration of their potential limitations regarding therapeutic translation. An enhanced understanding of treatment resistant depression is sorely needed given the burden of disease and lack of effective therapies.
Collapse
Affiliation(s)
- Julianna E Peña
- Department of Neurosurgery, University of Texas Southwestern Medical Center, Dallas, TX, United States; Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Brian F Corbett
- Department of Biology, Rutgers University, Camden, NJ, United States
| | - Carol A Tamminga
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Seema Bhatnagar
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, United States
| | - Frederick L Hitti
- Department of Neurosurgery, University of Texas Southwestern Medical Center, Dallas, TX, United States; Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| |
Collapse
|
3
|
Zou Z, Huang Y, Maes M, Wang J, He Y, Min W, Zhou B. Effects of antidepressant on FKBP51 mRNA expression and neuroendocrine hormones in patients with panic disorder. BMC Psychiatry 2024; 24:269. [PMID: 38600448 PMCID: PMC11005249 DOI: 10.1186/s12888-024-05704-4] [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: 07/02/2023] [Accepted: 03/21/2024] [Indexed: 04/12/2024] Open
Abstract
OBJECTIVE The purpose of this study was to investigate the effects of escitalopram on the peripheral expression of hypothalamic-pituitary-adrenal (HPA) axis-related genes (FKBP51, HSP90, NR3C1 and POMC) and HPA-axis hormones in patients with panic disorder (PD). METHODS Seventy-seven patients with PD were treated with escitalopram for 12 weeks. All participants were assessed for the severity of panic symptoms using the Panic Disorder Severity Scale (PDSS). The expression of HPA-axis genes was measured using real-time quantitative fluorescent PCR, and ACTH and cortisol levels were measured using chemiluminescence at baseline and after 12 weeks of treatment. RESULTS At baseline, patients with PD had elevated levels of ACTH and cortisol, and FKBP51 expression in comparison to healthy controls (all p < 0.01). Correlation analysis revealed that FKBP51 expression levels were significantly positively related to cortisol levels and the severity of PD (all p < 0.01). Furthermore, baseline ACTH and cortisol levels, and FKBP51 expression levels were significantly reduced after 12 weeks of treatment, and the change in the PDSS score from baseline to post-treatment was significantly and positively related to the change in cortisol (p < 0.01). CONCLUSIONS The results suggest that PD may be associated with elevated levels of ACTH and cortisol, and FKBP51 expression, and that all three biomarkers are substantially decreased in patients who have received escitalopram treatment.
Collapse
Affiliation(s)
- Zhili Zou
- Provincial Center for Mental Health, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, 610072, Chengdu, China.
| | - Yulan Huang
- Provincial Center for Mental Health, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, 610072, Chengdu, China
| | - Michael Maes
- Provincial Center for Mental Health, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, 610072, Chengdu, China
| | - Jinyu Wang
- Provincial Center for Mental Health, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, 610072, Chengdu, China
| | - Ying He
- Provincial Center for Mental Health, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, 610072, Chengdu, China
| | - Wenjiao Min
- Provincial Center for Mental Health, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, 610072, Chengdu, China
| | - Bo Zhou
- Provincial Center for Mental Health, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, 610072, Chengdu, China
| |
Collapse
|
4
|
Xiong SY, Wen HZ, Dai LM, Lou YX, Wang ZQ, Yi YL, Yan XJ, Wu YR, Sun W, Chen PH, Yang SZ, Qi XW, Zhang Y, Wu GY. A brain-tumor neural circuit controls breast cancer progression in mice. J Clin Invest 2023; 133:e167725. [PMID: 37847562 PMCID: PMC10721160 DOI: 10.1172/jci167725] [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: 12/05/2022] [Accepted: 10/12/2023] [Indexed: 10/18/2023] Open
Abstract
Tumor burden, considered a common chronic stressor, can cause widespread anxiety. Evidence suggests that cancer-induced anxiety can promote tumor progression, but the underlying neural mechanism remains unclear. Here, we used neuroscience and cancer tools to investigate how the brain contributes to tumor progression via nerve-tumor crosstalk in a mouse model of breast cancer. We show that tumor-bearing mice exhibited significant anxiety-like behaviors and that corticotropin-releasing hormone (CRH) neurons in the central medial amygdala (CeM) were activated. Moreover, we detected newly formed sympathetic nerves in tumors, which established a polysynaptic connection to the brain. Pharmacogenetic or optogenetic inhibition of CeMCRH neurons and the CeMCRH→lateral paragigantocellular nucleus (LPGi) circuit significantly alleviated anxiety-like behaviors and slowed tumor growth. Conversely, artificial activation of CeMCRH neurons and the CeMCRH→LPGi circuit increased anxiety and tumor growth. Importantly, we found alprazolam, an antianxiety drug, to be a promising agent for slowing tumor progression. Furthermore, we show that manipulation of the CeMCRH→LPGi circuit directly regulated the activity of the intratumoral sympathetic nerves and peripheral nerve-derived norepinephrine, which affected tumor progression by modulating antitumor immunity. Together, these findings reveal a brain-tumor neural circuit that contributes to breast cancer progression and provide therapeutic insights for breast cancer.
Collapse
Affiliation(s)
- Si-Yi Xiong
- Breast and Thyroid Surgery, Southwest Hospital
| | - Hui-Zhong Wen
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, College of Basic Medical Sciences
| | - Li-Meng Dai
- Department of Medical Genetics, College of Basic Medical Sciences
| | - Yun-Xiao Lou
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, College of Basic Medical Sciences
| | - Zhao-Qun Wang
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, College of Basic Medical Sciences
| | - Yi-Lun Yi
- Experimental Center of Basic Medicine, Chongqing Key Laboratory of Neurobiology, College of Basic Medical Sciences
| | - Xiao-Jing Yan
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences
| | - Ya-Ran Wu
- Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, and
| | - Wei Sun
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Peng-Hui Chen
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, College of Basic Medical Sciences
| | - Si-Zhe Yang
- Breast and Thyroid Surgery, Southwest Hospital
| | - Xiao-Wei Qi
- Breast and Thyroid Surgery, Southwest Hospital
| | - Yi Zhang
- Breast and Thyroid Surgery, Southwest Hospital
| | - Guang-Yan Wu
- Experimental Center of Basic Medicine, Chongqing Key Laboratory of Neurobiology, College of Basic Medical Sciences
| |
Collapse
|
5
|
Shi TS, Li WY, Chen YM, Huang J, Guan W, Xu DW, Jiang B. The antidepressant-like effects of escitalopram in mice require salt-inducible kinase 1 and CREB-regulated transcription co-activator 1 in the paraventricular nucleus of the hypothalamus. J Affect Disord 2023; 338:228-238. [PMID: 37257779 DOI: 10.1016/j.jad.2023.05.085] [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: 03/19/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
BACKGROUND The salt-inducible kinase 1 (SIK1)-CREB-regulated transcription co-activator 1 (CRTC1) system in the paraventricular nucleus (PVN) of the hypothalamus has been demonstrated to participate in not only depression neurobiology but also the antidepressant mechanisms of fluoxetine, paroxetine, venlafaxine, and duloxetine. Like fluoxetine and paroxetine, escitalopram is also a well-known selective serotonin (5-HT) reuptake inhibitor (SSRI). However, recently it has been found that escitalopram can modulate a lot of targets other than the 5-HT system. Here, we speculate that escitalopram produces effects on the SIK1-CRTC1 system in the PVN. METHODS Two mice models of depression (chronic social defeat stress (CSDS) and chronic unpredictable mild stress (CUMS)), various behavioral tests, enzyme linked immunosorbent assay (ELISA), western blotting, co-immunoprecipitation (Co-IP), quantitative real-time reverse transcription PCR (qRT-PCR), immunofluorescence, and adeno-associated virus (AAV)-mediated gene transfer were used together in the present study. RESULTS It was found that escitalopram administration not only significantly prevented the hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis induced by CSDS and CUMS, but also notably reversed the effects of CSDS and CUMS on SIK1, CRTC1, and CRTC1-CREB binding in the PVN of mice. AAV-based genetic knock-down of SIK1 in PVN neurons evidently abolished the antidepressant-like effects of escitalopram in mice. LIMITATION A shortage of this study is that only rodent models of depression were used, while human samples were not included. CONCLUSIONS In summary, regulating the SIK1-CRTC1 system in the PVN participates in the antidepressant mechanism of escitalopram, which extends the knowledge of the pharmacological actions of escitalopram.
Collapse
Affiliation(s)
- Tian-Shun Shi
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Wei-Yu Li
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Yan-Mei Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Jie Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Wei Guan
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Da-Wei Xu
- Department of Orthopedics, Affiliated Hospital 2 of Nantong University, Nantong 226006, Jiangsu, China.
| | - Bo Jiang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China.
| |
Collapse
|
6
|
Cui L, Zhang Q, Zhang Y, Li T, Li M, Yuan J, Wu Z, Zhang Y, Kong H, Qu H, Zhao Y. Anxiolytic effects of Chrysanthemum morifolium Ramat Carbonisata-based carbon dots in mCPP-induced anxiety-like behavior in mice: a nature-inspired approach. Front Mol Biosci 2023; 10:1222415. [PMID: 37520324 PMCID: PMC10373738 DOI: 10.3389/fmolb.2023.1222415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 06/26/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction: Anxiety disorders have emerged as a predominant health concern, yet existing pharmacological treatments for anxiety still present various challenges. Chrysanthemum morifolium Ramat Carbonisata (CMRC) has been utilized in China for approximately 400 years as a therapeutic intervention for anxiety disorders. In this study, a novel type of carbon dots derived from the decoction of Chrysanthemum morifolium Ramat Carbonisata (CMRC-CDs) was identified and isolated, and their morphological structure and functional groups were characterized. Furthermore, the effects of CMRC-CDs on m-chlorophenylpiperazine (mCPP)-induced anxiety-like behaviour in mice were examined and quantified. In order to investigate the potential mechanisms of their anxiolytic effects, concentrations of hypothalamic-pituitary-adrenal (HPA) axis hormones, amino acid neurotransmitters, and monoamine neurotransmitters were measured. Methods: In this study, we synthesized CMRC-CDs and evaluated their potential anti-anxiety effects in a controlled experiment involving 48 male ICR mice. The mice were randomly divided into six groups, treated with CMRC-CDs at different doses for 14 days, and subjected to Open-Field (OF) and Elevated Plus Maze (EPM) tests. Post-behavioral evaluations, blood samples and brain tissues were collected for neurotransmitter and Hypothalamic-Pituitary-Adrenal (HPA) axis hormone quantification via ELISA. Additionally, cytotoxicity of CMRC-CDs was assessed using a Cell Counting Kit-8 (CCK-8) assay on RAW 264.7 cells. Results and Discussion: CMRC-CDs were spherical and homogeneously dispersed, with diameters ranging from 1.4 to 4.0 nm and an abundance of chemical groups on their surface. In the open-field (OF) test, mice pre-treated with CMRC-CDs demonstrated an increased proportion of time spent in the central area and a higher frequency of entries into the central area. In the elevated plus maze (EPM) test, mice pre-treated with CMRC-CDs exhibited a greater number of entries into the open arm and an extended duration spent in the open arm. CMRC-CDs were observed to decrease serum concentrations of corticotropin-releasing hormone (CRH), adrenocorticotropic hormone (ACTH), and corticosterone (CORT). Furthermore, CMRC-CDs were found to increase γ-aminobutyric acid (GABA) and 5-hydroxytryptamine (5-HT) levels, while concurrently reducing glutamic acid (Glu) concentrations in brain tissue. CMRC-CDs demonstrated anxiolytic effects, which may be attributed to their modulation of hormones and neurotransmitters. This finding suggests the potential therapeutic value of CMRC-CDs in the clinical treatment of anxiety disorders.
Collapse
Affiliation(s)
- Luming Cui
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qian Zhang
- Qingdao Zhonghengneng Environmental Science Engineering Research Institute Co., Ltd, Qingdao, China
| | - Yifan Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tingjie Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Menghan Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jinye Yuan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhiyi Wu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yue Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Hui Kong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Huihua Qu
- Center of Scientific Experiment, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
7
|
Islas-Preciado D, López-Rubalcava C, Estrada-Camarena E, de Gortari P, Castro-García M. Effect of chronic unpredictable stress in female Wistar-Kyoto rats subjected to progesterone withdrawal: Relevance for Premenstrual Dysphoric Disorder neurobiology. Psychoneuroendocrinology 2023; 155:106331. [PMID: 37437420 DOI: 10.1016/j.psyneuen.2023.106331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/14/2023]
Abstract
Premenstrual Dysphoric Disorder (PMDD) is related to an abrupt drop in progesterone and impairments in the HPA axis that cause anxiety. Suffering persons report higher daily-life stress and anxiety proneness that may contribute to developing PMDD, considered a chronic stress-related disorder. Here, we explored the effect of chronic unpredictable stress (CUS) in rats subjected to progesterone withdrawal (PW) and evaluated gene expression of HPA axis activation in the stress-vulnerable Wistar-Kyoto (WKY) rat strain that is prone to anxiety. Ovariectomized WKY rats were randomly assigned to CUS or Standard-housed conditions (SHC) for 30 days. To induce PW, animals received 2 mg/kg of progesterone on day 25th for 5 days; 24 h later, they were tested using the anxiety-like burying behavior test (BBT). After behavioral completion, rats were euthanized, and brains were extracted to measure Crh (PVN) and Nr3c1 (hippocampus) mRNA. Blood corticosterone and vasopressin levels were determined. Results showed that PW exacerbated anxiety-like behaviors through passive coping in CUS-WKY. PW decreased Crh-PVN mRNA and the Nr3c1-hippocampal mRNA expression in SHC. CUS decreased Crh-PVN mRNA compared to SHC, and no further changes were observed by PW or BBT exposure. CUS reduced Nr3c1-hippocampal gene expression compared to SHC animals, and lower Nr3c1 mRNA was detected due to BBT. The PW increased corticosterone in SHC and CUS rats; however, CUS blunted corticosterone when combined with PW+BBT and similarly occurred in vasopressin concentrations. Chronic stress blunts the response of components of the HPA axis regulation when PW and BBT (systemic and psychogenic stressors, respectively) are presented. This response may facilitate less adaptive behaviors through passive coping in stress-vulnerable subjects in a preclinical model of premenstrual anxiety.
Collapse
Affiliation(s)
- D Islas-Preciado
- Lab. de Neuropsicofarmacología, Dirección de Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico
| | - C López-Rubalcava
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados IPN (Cinvestav-IPN), Mexico
| | - E Estrada-Camarena
- Lab. de Neuropsicofarmacología, Dirección de Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico.
| | - P de Gortari
- Lab. de Neurofisiología Molecular, Dirección de Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico
| | - M Castro-García
- Lab de Etología, Dirección de Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico
| |
Collapse
|
8
|
Gołyszny M, Zieliński M, Paul-Samojedny M, Pałasz A, Obuchowicz E. Chronic treatment with escitalopram and venlafaxine affects the neuropeptide S pathway differently in adult Wistar rats exposed to maternal separation. AIMS Neurosci 2022; 9:395-422. [PMID: 36329901 PMCID: PMC9581731 DOI: 10.3934/neuroscience.2022022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/20/2022] [Accepted: 09/05/2022] [Indexed: 07/05/2024] Open
Abstract
Neuropeptide S (NPS), which is a peptide that is involved in the regulation of the stress response, seems to be relevant to the mechanism of action of antidepressants that have anxiolytic properties. However, to date, there have been no reports regarding the effect of long-term treatment with escitalopram or venlafaxine on the NPS system under stress conditions. This study aimed to investigate the effects of the above-mentioned antidepressants on the NPS system in adult male Wistar rats that were exposed to neonatal maternal separation (MS). Animals were exposed to MS for 360 min. on postnatal days (PNDs) 2-15. MS causes long-lasting behavioral, endocrine and neurochemical consequences that mimic anxiety- and depression-related features. MS and non-stressed rats were given escitalopram or venlafaxine (10mg/kg) IP from PND 69 to 89. The NPS system was analyzed in the brainstem, hypothalamus, amygdala and anterior olfactory nucleus using quantitative RT-PCR and immunohistochemical methods. The NPS system was vulnerable to MS in the brainstem and amygdala. In the brainstem, escitalopram down-regulated NPS and NPS mRNA in the MS rats and induced a tendency to reduce the number of NPS-positive cells in the peri-locus coeruleus. In the MS rats, venlafaxine insignificantly decreased the NPSR mRNA levels in the amygdala and a number of NPSR cells in the basolateral amygdala, and increased the NPS mRNA levels in the hypothalamus. Our data show that the studied antidepressants affect the NPS system differently and preliminarily suggest that the NPS system might partially mediate the pharmacological effects that are induced by these drugs.
Collapse
Affiliation(s)
- Miłosz Gołyszny
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland
| | - Michał Zieliński
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland
| | - Monika Paul-Samojedny
- Department of Medical Genetics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland
| | - Artur Pałasz
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland
| | - Ewa Obuchowicz
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland
| |
Collapse
|
9
|
Gołyszny M, Zieliński M, Paul-Samojedny M, Filipczyk Ł, Pałasz A, Obuchowicz E. Escitalopram alters the hypothalamic OX system but does not affect its up-regulation induced by early-life stress in adult rats. Neurosci Res 2022; 180:58-71. [PMID: 35219722 DOI: 10.1016/j.neures.2022.02.005] [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: 07/21/2021] [Revised: 02/07/2022] [Accepted: 02/23/2022] [Indexed: 01/06/2023]
Abstract
We hypothesized that there is a relationship between the orexinergic system (OX) alterations and changes elicited by escitalopram or venlafaxine in adult rats subjected to maternal separation (MS). This animal model of childhood adversity induces long-lasting consequences in adult physiology and behavior. Male Wistar rats from the control and MS groups were injected with escitalopram or venlafaxine (10 mg/kg) IP from postnatal day (PND) 69-89. Adult rats were subjected to behavioral assessment, estimation of hypothalamic-pituitary-adrenal (HPA) axis activity and analysis of the OX system (quantitative PCR and immunohistochemistry) in the hypothalamus and amygdala. MS caused anxiety- and depressive-like behavior, endocrine stress-related response, and up-regulation of the OX system in the hypothalamus. Escitalopram, but not venlafaxine, increased the activity of hypothalamic OX system in the control rats and both drugs had no effect on OXs in the MS group. The disturbed signaling of the OX pathway may be significant for harmful long-term consequences of early-life stress. Our data show that the normal brain and brain altered by MS respond differently to escitalopram. Presumably, anti-anxiety and antidepressant effects of this drug do not depend on the activity of hypothalamic OX system.
Collapse
Affiliation(s)
- Miłosz Gołyszny
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18 street, Katowice 40-752, Poland.
| | - Michał Zieliński
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18 street, Katowice 40-752, Poland
| | - Monika Paul-Samojedny
- Department of Medical Genetics, Faculty of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Jedności 8, Sosnowiec 41-200, Poland
| | - Łukasz Filipczyk
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18 street, Katowice 40-752, Poland
| | - Artur Pałasz
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18 street, Katowice 40-752, Poland
| | - Ewa Obuchowicz
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18 street, Katowice 40-752, Poland
| |
Collapse
|
10
|
Zhou L, Wang T, Yu Y, Li M, Sun X, Song W, Wang Y, Zhang C, Fu F. The etiology of poststroke-depression: a hypothesis involving HPA axis. Biomed Pharmacother 2022; 151:113146. [PMID: 35643064 DOI: 10.1016/j.biopha.2022.113146] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/06/2022] [Accepted: 05/15/2022] [Indexed: 11/30/2022] Open
Abstract
Approximately, one in three ischemic stroke survivors suffered from depression, namely, post-stroke depression (PSD). PSD affects functional rehabilitation and may lead to poor quality of life of patients. There are numerous explanations about the etiologies of PSD. Here, we speculated that PSD are likely to be the result of specific changes in brain pathology. We hypothesized that the stroke-induced hyperactivity of hypothalamic-pituitary-adrenal (HPA) axis plays an important role in PSD. Stroke initiates a complex sequence of events in neuroendocrine system including HPA axis. The HPA axis is involved in the pathophysiology of depression, especially, the overactivity of the HPA axis occurs in major depressive disorder. This review summarizes the possible etiologies of PSD, focusing on the stroke-induced activation of HPA axis, mainly including the stress followed by severe brain damage and the proinflammatory cytokines release. The role of hyperactive of HPA axis in PSD was discussed in detail, which includes the role of high level corticotropin-releasing hormone in PSD, the effects of glucocorticoids on the alterations in specific brain structures, the expression of enzymes, excitotoxicity, the change in intestinal permeability, and the activation of microglia. The relationship between neuroendocrine regulation and inflammation was also described. Finally, the therapy of PSD by regulating HPA axis, neuroendocrine, and immunity was discussed briefly. Nevertheless, the change of HPA axis and the occurring of PSD maybe interact and promote on each other, and future investigations should explore this hypothesis in more depth.
Collapse
Affiliation(s)
- Lin Zhou
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Tian Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Yawen Yu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Mingan Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Xiaohui Sun
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Wenhao Song
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Yunjie Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Ce Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Fenghua Fu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China.
| |
Collapse
|
11
|
Piwowarczyk-Nowak A, Pałasz A, Bogus K, Krzystanek M, Błaszczyk I, Worthington JJ, Grajoszek A. Modulatory effect of long-term treatment with escitalopram and clonazepam on the expression of anxiety-related neuropeptides: neuromedin U, neuropeptide S and their receptors in the rat brain. Mol Biol Rep 2022; 49:9041-9049. [PMID: 35690686 DOI: 10.1007/s11033-022-07578-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/17/2022] [Accepted: 05/06/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Newly identified multifunctional peptidergic modulators of stress responses: neuromedin U (NMU) and neuropeptide S (NPS) are involved in the wide spectrum of brain functions. However, there are no reports dealing with potential molecular relationships between the action of diverse anxiolytic or antidepressant drugs and NMU and NPS signaling in the brain. The present work was therefore focused on local expression of the aforementioned stress-related neuropeptides in the rat brain after long-term treatment with escitalopram and clonazepam. METHODS Studies were carried out on adult, male Sprague-Dawley rats that were divided into 3 groups: animals injected with saline (control) and experimental individuals treated with escitalopram (at single dose 5 mg/kg daily), and clonazepam (at single dose 0.5 mg/kg). All individuals were sacrificed under anaesthesia and the whole brain excised. Total mRNA was isolated from homogenized samples of amygdala, hippocampus, hypothalamus, thalamus, cerebellum and brainstem. Real time-PCR method was used for estimation of related NPS, NPS receptor (NPSR), NMU, NMU and receptor 2 (NMUR2) mRNA expression. The whole brains were also sliced for general immunohistochemical assessment of the neuropeptides expression. RESULTS Chronic administration of clonazepam resulted in an increase of NMU mRNA expression and formation of NMU-expressing fibers in the amygdala, while escitalopram produced a significant decrease in NPSR mRNA level in hypothalamus. Long-term escitalopram administration affects the local expression of examined neuropeptides mRNA in a varied manner depending on the brain structure. CONCLUSIONS Pharmacological effects of escitalopram may be connected with local at least partially NPSR-related alterations in the NPS/NMU/NMUR2 gene expression at the level selected rat brain regions. A novel alternative mode of SSRI action can be therefore cautiously proposed.
Collapse
Affiliation(s)
- Aneta Piwowarczyk-Nowak
- Department of Anatomy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland
| | - Artur Pałasz
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland.
| | - Katarzyna Bogus
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland
| | - Marek Krzystanek
- Clinic of Psychiatric Rehabilitation, Department of Psychiatry and Psychotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Ziolowa 45/47 Katowice 40- 635, Katowice, Poland
| | - Iwona Błaszczyk
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland
| | - John J Worthington
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, LA1 4YQ, Lancaster, UK
| | - Aniela Grajoszek
- Department for Experimental Medicine, Medical University of Silesia, ul. Medyków 4, 40-752, Katowice, Poland
| |
Collapse
|
12
|
Abdelwahab LA, Galal OO, Abd El-Rahman SS, El-Brairy AI, Khattab MM, El-Khatib AS. Targeting the oxytocin system to ameliorate early life depressive-like behaviors in maternally-separated rats. Biol Pharm Bull 2021; 44:1445-1457. [PMID: 34349049 DOI: 10.1248/bpb.b21-00247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Oxytocin (OXT) - "the love hormone" - has been involved in the anti-depressant activity of some selective serotonin reuptake inhibitors (SSRIs). The exact mechanism underlying the OXT pathway in depression is not fully clear. This study aimed to investigate the effect of OXT analogue, carbetocin (CBT) and the SSRI, escitalopram (ESCIT) on depressive-like behaviors following maternal separation (MS). It is worthy to mention that intranasal CBT has been approved by FDA for Prader-Willi syndrome. Adolescent Wistar albino maternally-separated rats were given CBT, (100 μg/animal/day via inhalation route), and, ESCIT, (20 mg kg-1, po) either alone or in combination for 7 days. Repeated 3-h MS demonstrated increased immobility time in forced swim test (FST) and decreased locomotor activity in open field test. MS elevated plasma level of adrenocortico-trophic hormone (ACTH) but notably reduced plasma OXT, with no effect on hippocampal OXT-R expression. Following MS, hippocampal contents of 5-hydroxytryptamine receptors (5HT1A-R), serotonin transporter (SERT) were increased. CBT and ESCIT corrected the behavioral dysfunction in FST and suppressed the high levels of ACTH. Additionally, both treatments boosted OXT level, reduced 5HT1A-R and normalized SERT contents, which reflects increased availability of serotonin. Finally, CBT markedly ameliorated the histopathological damage induced by MS and suppressed the increased glial fibrillary acidic protein. CBT and ESCIT manage depressive-like behavior by positively affecting serotonergic and oxytocinergic systems. Targeting OXT system -using CBT- ameliorated depressive like behaviors induced by maternal separation most probably via enhancing OXT plasma levels, attenuating hormonal ACTH and restoring the expression of hippocampal oxytocin and serotonin mechanisms.
Collapse
Affiliation(s)
- Lobna A Abdelwahab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA)
| | - Omneya O Galal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University (ACU)
| | | | - Amany I El-Brairy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA)
| | - Mahmoud M Khattab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University
| | - Aiman S El-Khatib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University
| |
Collapse
|
13
|
Murata S, Murphy M, Khanna R, Hoppensteadt D, Fareed J, Halaris A. Elevated salivary cortisol predicts response to adjunctive immune modulation in treatment-resistant bipolar depression. JOURNAL OF AFFECTIVE DISORDERS REPORTS 2021. [DOI: 10.1016/j.jadr.2021.100117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
14
|
Henn L, Zanta NC, Girardi CEN, Suchecki D. Chronic Escitalopram Treatment Does Not Alter the Effects of Neonatal Stress on Hippocampal BDNF Levels, 5-HT 1A Expression and Emotional Behaviour of Male and Female Adolescent Rats. Mol Neurobiol 2021; 58:926-943. [PMID: 33063280 DOI: 10.1007/s12035-020-02164-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/07/2020] [Indexed: 10/23/2022]
Abstract
Early life stress is considered a risk factor for the development of long-term psychiatric disorders. Maternal deprivation (MD) is a useful paradigm to understand the neurobiological underpinnings of early stress-induced changes in neurodevelopment trajectory. The goal of the present study was to examine the effects of a chronic treatment with escitalopram (ESC) on the hippocampal levels of BDNF and neuropeptide Y (NPY), expression of serotonin type 1A receptor (5-HT1A), plasma corticosterone levels and emotional behaviours in male and female adolescent rats submitted to MD at 9 days of life (group DEP9) and challenged with a brief and mild stress (saline injection (SAL)) at the end of MD. Whole litters were kept with mothers (CTL) or submitted to MD (DEP9). Within each group, pups were stress-challenged (CTL-SAL and DEP9-SAL) or not (CTL-NSAL and DEP9-NSAL). ESC or vehicle treatments began at weaning and lasted 24 days, when animals were sacrificed for determination of neurobiological variables or submitted to a battery of tests for evaluation of emotional behaviours. The results showed that BDNF levels were higher in SAL-challenged males and in DEP9-SAL females, whereas 5-HT1A receptor expression was reduced in DEP9 males and in SAL-challenged females. There were no changes in NPY or corticosterone levels. In the forced swim test, SAL-challenged males and DEP9 females displayed less immobility and ESC only increased social motivation in males. The results indicated that neonatal stress led to sex-dependent changes in neurobiology and behaviour and that chronic ESC treatment had minor effects on these parameters.
Collapse
Affiliation(s)
- Lorena Henn
- Departamento de Psicobiologia, Universidade Federal de São Paulo, Rua Napoleão de Barros, 925, 1° andar, São Paulo, SP, 04024-002, Brazil
| | - Natália C Zanta
- Departamento de Psicobiologia, Universidade Federal de São Paulo, Rua Napoleão de Barros, 925, 1° andar, São Paulo, SP, 04024-002, Brazil
| | - Carlos Eduardo N Girardi
- Departamento de Psicobiologia, Universidade Federal de São Paulo, Rua Napoleão de Barros, 925, 1° andar, São Paulo, SP, 04024-002, Brazil
| | - Deborah Suchecki
- Departamento de Psicobiologia, Universidade Federal de São Paulo, Rua Napoleão de Barros, 925, 1° andar, São Paulo, SP, 04024-002, Brazil.
| |
Collapse
|
15
|
Han YM, Kim MS, Jo J, Shin D, Kwon SH, SEO JB, Kang D, Lee BD, Ryu H, Hwang EM, Kim JM, Patel PD, Lyons DM, Schatzberg AF, Her S. Decoding the temporal nature of brain GR activity in the NFκB signal transition leading to depressive-like behavior. Mol Psychiatry 2021; 26:5087-5096. [PMID: 33483691 PMCID: PMC7821461 DOI: 10.1038/s41380-021-01016-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 11/17/2020] [Accepted: 01/05/2021] [Indexed: 01/30/2023]
Abstract
The fine-tuning of neuroinflammation is crucial for brain homeostasis as well as its immune response. The transcription factor, nuclear factor-κ-B (NFκB) is a key inflammatory player that is antagonized via anti-inflammatory actions exerted by the glucocorticoid receptor (GR). However, technical limitations have restricted our understanding of how GR is involved in the dynamics of NFκB in vivo. In this study, we used an improved lentiviral-based reporter to elucidate the time course of NFκB and GR activities during behavioral changes from sickness to depression induced by a systemic lipopolysaccharide challenge. The trajectory of NFκB activity established a behavioral basis for the NFκB signal transition involved in three phases, sickness-early-phase, normal-middle-phase, and depressive-like-late-phase. The temporal shift in brain GR activity was differentially involved in the transition of NFκB signals during the normal and depressive-like phases. The middle-phase GR effectively inhibited NFκB in a glucocorticoid-dependent manner, but the late-phase GR had no inhibitory action. Furthermore, we revealed the cryptic role of basal GR activity in the early NFκB signal transition, as evidenced by the fact that blocking GR activity with RU486 led to early depressive-like episodes through the emergence of the brain NFκB activity. These results highlight the inhibitory action of GR on NFκB by the basal and activated hypothalamic-pituitary-adrenal (HPA)-axis during body-to-brain inflammatory spread, providing clues about molecular mechanisms underlying systemic inflammation caused by such as COVID-19 infection, leading to depression.
Collapse
Affiliation(s)
- Young-Min Han
- grid.410885.00000 0000 9149 5707Seoul Centre, Korea Basic Science Institute, Seoul, South Korea
| | - Min Sun Kim
- grid.410885.00000 0000 9149 5707Seoul Centre, Korea Basic Science Institute, Seoul, South Korea
| | - Juyeong Jo
- grid.410885.00000 0000 9149 5707Seoul Centre, Korea Basic Science Institute, Seoul, South Korea
| | - Daiha Shin
- grid.410885.00000 0000 9149 5707Seoul Centre, Korea Basic Science Institute, Seoul, South Korea
| | - Seung-Hae Kwon
- grid.410885.00000 0000 9149 5707Seoul Centre, Korea Basic Science Institute, Seoul, South Korea
| | - Jong Bok SEO
- grid.410885.00000 0000 9149 5707Seoul Centre, Korea Basic Science Institute, Seoul, South Korea
| | - Dongmin Kang
- grid.255649.90000 0001 2171 7754Department of Life Science, Ewha Womans University, Seoul, South Korea
| | - Byoung Dae Lee
- grid.289247.20000 0001 2171 7818Department of Physiology, School of Medicine, Kyung Hee University, Seoul, South Korea
| | - Hoon Ryu
- grid.35541.360000000121053345Neuroscience Centre, Korea Institute of Science and Technology, Seoul, South Korea
| | - Eun Mi Hwang
- grid.35541.360000000121053345Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul, South Korea
| | - Jae-Min Kim
- grid.14005.300000 0001 0356 9399Department of Psychiatry, Chonnam National University Medical School, Seoul, South Korea
| | - Paresh D. Patel
- grid.412590.b0000 0000 9081 2336Department of Psychiatry, Molecular and Behavioral Neuroscience Institute, University of Michigan Medical Centre, Ann Arbor, MI USA
| | - David M. Lyons
- grid.168010.e0000000419368956Departments of Psychiatry, Stanford University Medical Centre, Stanford, CA USA
| | - Alan F. Schatzberg
- grid.168010.e0000000419368956Departments of Psychiatry, Stanford University Medical Centre, Stanford, CA USA
| | - Song Her
- Seoul Centre, Korea Basic Science Institute, Seoul, South Korea.
| |
Collapse
|
16
|
Escitalopram as a modulator of proopiomelanocortin, kisspeptin, Kiss1R and MCHR1 gene expressions in the male rat brain. Mol Biol Rep 2020; 47:8273-8278. [PMID: 32914264 PMCID: PMC7588374 DOI: 10.1007/s11033-020-05806-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/03/2020] [Indexed: 10/24/2022]
Abstract
Neuropeptides are important, multifunctional regulatory factors of the nervous system, being considered as a novel, atypical sites of antidepressants action. It has already been proven that some of them, such as selective serotonin reuptake inhibitors (SSRI), are able to affect peptidergic pathways in various brain regions. Despite these reports, there is so far no reports regarding the effect of treatment with SSRIs on brain proopiomelanocortin (POMC), kisspeptin, Kiss1R and MCHR1 gene expression. In the current study we examined POMC, kisspeptin, Kiss1R and MCHR1 mRNA expression in the selected brain structures (hypothalamus, hippocampus, amygdala, striatum, cerebellum and brainstem) of rats chronically treated with a 10 mg/kg dose of escitalopram using quantitative Real-Time PCR. Long-term treatment with escitalopram led to the upregulation of MCHR1 expression in the rat amygdala. Kisspeptin mRNA level was also increased in the amygdala, but Kiss1R mRNA expressions were elevated in the hippocampus, hypothalamus and cerebellum. POMC mRNA expressions were in turn decreased in the hippocampus, amygdala, cerebellum and brainstem. These results may support the hypothesis that these neuropeptides may be involved in the site-dependent actions of SSRI antidepressants. This is the first report of the effects of escitalopram on POMC, kisspeptin, Kiss1R and MCHR1 in animal brain. Our findings shed a new light on the pharmacology of SSRIs and may contribute to a better understanding of the alternative, neuropeptide-dependent modes of antidepressant action.
Collapse
|
17
|
Tafet GE, Nemeroff CB. Pharmacological Treatment of Anxiety Disorders: The Role of the HPA Axis. Front Psychiatry 2020; 11:443. [PMID: 32499732 PMCID: PMC7243209 DOI: 10.3389/fpsyt.2020.00443] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/01/2020] [Indexed: 12/13/2022] Open
Abstract
Stress in general, and early life stress in particular, has been associated with the development of anxiety and mood disorders. The molecular, biological and psychological links between stress exposure and the pathogenesis of anxiety and mood disorders have been extensively studied, resulting in the search of novel psychopharmacological strategies aimed at targets of the hypothalamic-pituitary-adrenal (HPA) axis. Hyperactivity of the HPA axis has been observed in certain subgroups of patients with anxiety and mood disorders. In addition, the effects of different anti-anxiety agents on various components of the HPA axis has been investigated, including benzodiazepines, tricyclic antidepressants (TCAs), and selective serotonin reuptake inhibitors (SSRIs). For example, benzodiazepines, including clonazepam and alprazolam, have been demonstrated to reduce the activity of corticotrophin releasing factor (CRF) neurons in the hypothalamus. TCAs and SSRIs are also effective anti-anxiety agents and these may act, in part, by modulating the HPA axis. In this regard, the SSRI escitalopram inhibits CRF release in the central nucleus of the amygdala, while increasing glucocorticoid receptor (GRs) density in the hippocampus and hypothalamus. The molecular effects of these anti-anxiety agents in the regulation of the HPA axis, taken together with their clinical efficacy, may provide further understanding about the role of the HPA axis in the pathophysiology of mood and anxiety disorders, paving the way for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Gustavo E. Tafet
- Department of Psychiatry and Neurosciences, Maimónides University, Buenos Aires, Argentina
| | - Charles B. Nemeroff
- Department of Psychiatry, University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
18
|
Perinatal exposure to venlafaxine leads to lower anxiety and depression-like behavior in the adult rat offspring. Behav Pharmacol 2019; 29:445-452. [PMID: 29561291 DOI: 10.1097/fbp.0000000000000393] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Depression during pregnancy and in the post-partum period is a growing health issue. Venlafaxine, a representative of serotonin and noradrenaline reuptake inhibitors, is used to treat a wide spectrum of mood disorders. However, the limited number of prenatal and perinatal studies raises the question about the long-term consequences of venlafaxine therapy. The aim of this study was to investigate the effect of venlafaxine exposure during pregnancy and lactation on anxiety-like and depression-like behaviors, as well as adrenocortical hormone concentrations in the adult rat offspring. For this purpose, rat dams were treated orally with venlafaxine from day 15 of gestation to postnatal day 20 at doses of 7.5, 37.5, and 75 mg/kg. Administration of venlafaxine during gestation and lactation affected anxiety-like and depression-like behaviors in adult rat offspring of both sexes. The animals exposed through their mothers to venlafaxine, particularly at the lowest and middle doses, were less anxious and less depressive in several relevant behavioral tests, which can be considered a deviation from the normal state. At clinically relevant doses, venlafaxine did not alter circulating level of corticosterone and aldosterone in the adult offspring. In general, the consequences of venlafaxine were dose dependent and more apparent in females. Together, these results suggest that prenatal and early postnatal exposure to venlafaxine may interfere with functional development of the brain, though not necessarily in a negative way.
Collapse
|
19
|
McNeal N, Watanasriyakul WT, Normann MC, Akinbo OI, Dagner A, Ihm E, Wardwell J, Grippo AJ. The negative effects of social bond disruption are partially ameliorated by sertraline administration in prairie voles. Auton Neurosci 2019; 219:5-18. [PMID: 31122602 PMCID: PMC6540807 DOI: 10.1016/j.autneu.2019.03.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 03/13/2019] [Accepted: 03/14/2019] [Indexed: 12/11/2022]
Abstract
Negative social experiences influence both depression and cardiovascular dysfunction. Many individuals who experience negative mood states or cardiovascular conditions have limited social support. Therefore, investigation of drug treatments that may protect against the consequences of social stress will aid in designing effective treatment strategies. The current study used an animal model to evaluate the protective effect of sertraline administration on behavioral and cardiovascular consequences of social stress. Specifically, male prairie voles (Microtus ochrogaster), which are socially monogamous rodents that share several behavioral and physiological characteristics with humans, were isolated from a socially-bonded female partner, and treated with sertraline (16 mg/kg/day, ip) or vehicle during isolation. Unexpectedly, sertraline did not protect against depression-relevant behaviors, and it was associated with increased short- and long-term heart rate responses. However, sertraline administration improved heart rate variability recovery following a behavioral stressor, including increased parasympathetic regulation, and altered long-term neuronal activity in brain regions that modulate autonomic control and stress reactivity. These results indicate that sertraline may partially protect against the consequences of social stressors, and suggest a mechanism through which sertraline may beneficially influence neurobiological control of cardiac function.
Collapse
Affiliation(s)
- Neal McNeal
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | | | - Marigny C Normann
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Oreoluwa I Akinbo
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Ashley Dagner
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Elliott Ihm
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Joshua Wardwell
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Angela J Grippo
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA.
| |
Collapse
|
20
|
Gołyszny M, Obuchowicz E. Are neuropeptides relevant for the mechanism of action of SSRIs? Neuropeptides 2019; 75:1-17. [PMID: 30824124 DOI: 10.1016/j.npep.2019.02.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 01/08/2019] [Accepted: 02/13/2019] [Indexed: 12/12/2022]
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are drugs of first choice in the therapy of moderate to severe depression and anxiety disorders. Their primary mechanism of action is via influence of the serotonergic (5-HT) system, but a growing amount of data provides evidence for other non-monoaminergic players in SSRI effects. It is assumed that neuropeptides, which play a role as neuromodulators in the CNS, are involved in their mechanism of action. In this review we focus on six neuropeptides: corticotropin-releasing factor - CRF, galanin - GAL, oxytocin - OT, vasopressin - AVP, neuropeptide Y - NPY, and orexins - OXs. First, information about their roles in depression and anxiety disorders are presented. Then, findings describing their interactions with the 5-HT system are summarized. These data provide background for analysis of the results of published preclinical and clinical studies related to SSRI effects on the neuropeptide systems. We also report findings showing how modulation of neuropeptide transmission influences behavioral and neurochemical effects of SSRIs. Finally, future research necessary for enriching our knowledge of SSRI mechanisms of action is proposed. Recognition of new molecular targets for antidepressants will have a significant effect on the development of novel therapeutic strategies for mood-related disorders.
Collapse
Affiliation(s)
- Miłosz Gołyszny
- Department of Pharmacology, School of Medicine in Katowice, Medical University of Silesia, Poniatowskiego 15, 40-055 Katowice, Poland
| | - Ewa Obuchowicz
- Department of Pharmacology, School of Medicine in Katowice, Medical University of Silesia, Poniatowskiego 15, 40-055 Katowice, Poland.
| |
Collapse
|
21
|
SSRIs and SNRIs (SRI) in Pregnancy: Effects on the Course of Pregnancy and the Offspring: How Far Are We from Having All the Answers? Int J Mol Sci 2019; 20:ijms20102370. [PMID: 31091646 PMCID: PMC6567187 DOI: 10.3390/ijms20102370] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 12/31/2022] Open
Abstract
Serotonin has important roles in the development of the brain and other organs. Manipulations of synaptic serotonin by drugs such as serotonin reuptake inhibitors (SRI) or serotonin norepinephrine reuptake inhibitors (SNRI) might alter their development and function. Of interest, most studies on the outcome of prenatal exposure to SRI in human have not found significant embryonic or fetal damage, except for a possible, slight increase in cardiac malformations. In up to a third of newborns exposed to SRI, exposure may induce transient neonatal behavioral changes (poor neonatal adaptation) and increased rate of persistent pulmonary hypertension. Prenatal SRI may also cause slight motor delay and language impairment but these are transient. The data on the possible association of prenatal SRIs with autism spectrum disorder (ASD) are inconsistent, and seem to be related to pre-pregnancy treatment or to maternal depression. Prenatal SRIs also appear to affect the hypothalamic hypophyseal adrenal (HPA) axis inducing epigenetic changes, but the long-term consequences of these effects on humans are as yet unknown. SRIs are metabolized in the liver by several cytochrome P450 (CYP) enzymes. Faster metabolism of most SRIs in late pregnancy leads to lower maternal concentrations, and thus potentially to decreased efficacy which is more prominent in women that are rapid metabolizers. Studies suggest that the serotonin transporter SLC6A4 promoter is associated with adverse neonatal outcomes after SRI exposure. Since maternal depression may adversely affect the child's development, one has to consider the risk of SRI discontinuation on the fetus and the child. As with any drug treatment in pregnancy, the benefits to the mother should be considered versus the possible hazards to the developing embryo/fetus.
Collapse
|
22
|
Wu C, Gong WG, Wang YJ, Sun JJ, Zhou H, Zhang ZJ, Ren QG. Escitalopram alleviates stress-induced Alzheimer's disease-like tau pathologies and cognitive deficits by reducing hypothalamic-pituitary-adrenal axis reactivity and insulin/GSK-3β signal pathway activity. Neurobiol Aging 2018; 67:137-147. [DOI: 10.1016/j.neurobiolaging.2018.03.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 02/10/2018] [Accepted: 03/10/2018] [Indexed: 10/17/2022]
|
23
|
Benatti C, Alboni S, Blom JMC, Mendlewicz J, Tascedda F, Brunello N. Molecular changes associated with escitalopram response in a stress-based model of depression. Psychoneuroendocrinology 2018; 87:74-82. [PMID: 29049934 DOI: 10.1016/j.psyneuen.2017.10.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/15/2017] [Accepted: 10/12/2017] [Indexed: 11/30/2022]
Abstract
Converging evidence points at hypothalamus-pituitary-adrenal (HPA) axis hyperactivity and neuroinflammation as important factors involved in the etiopathogenesis of major depressive disorder (MDD) and in therapeutic efficacy of antidepressants. In this study, we examined the molecular effects associated with a response to a week-long treatment with escitalopram in the chronic escape deficit (CED) model, a validated model of depression based on the induction of an escape deficit after exposure of rats to an unavoidable stress. We confirmed our previous result that a treatment with escitalopram (10mg/kg) was effective after 7days in reverting the stress-induced escape deficit in approximately 50% of the animals, separating responders from non-responders. Expression of markers of HPA axis functionality as well as several inflammatory mediators were evaluated in the hypothalamus, a key structure integrating signals from the neuro, immune, endocrine systems. In the hypothalamus of responder animals we observed a decrease in the expression of CRH and its receptors and an increase in GR protein in total and nuclear extracts; this effect was accompanied by a significant decrease in circulating corticosterone in the same cohort. Hypothalamic IL-1β and TNFα expression were increased in stressed animals, while CXCL2, IL-6, and ADAM17 mRNA levels were decreased in escitalopram treated rats regardless of the treatment response. These data suggest that efficacy of a one week treatment with escitalopram may be partially mediated by a decrease HPA axis activity, while in the hypothalamus the drug-induced effects on the expression of immune modulators did not correlate with the behavioural outcome.
Collapse
Affiliation(s)
- Cristina Benatti
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy; Center for Neuroscience and Neurotechnology University of Modena and Reggio Emilia, Modena, Italy.
| | - Silvia Alboni
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy
| | - Joan M C Blom
- Center for Neuroscience and Neurotechnology University of Modena and Reggio Emilia, Modena, Italy; Department of Education and Human Sciences, University of Modena and Reggio Emilia, Viale Antonio Allegri 9, 42121, Reggio Emilia, Italy
| | - Julien Mendlewicz
- Department of Psychiatry, University Clinics of Brussels, Erasme Hospital, Free University of Brussels, 808 Route de Lennik, Brussels, Belgium
| | - Fabio Tascedda
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy; Center for Neuroscience and Neurotechnology University of Modena and Reggio Emilia, Modena, Italy
| | - Nicoletta Brunello
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy; Center for Neuroscience and Neurotechnology University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
24
|
Neurobehavioral risks of SSRIs in pregnancy: Comparing human and animal data. Reprod Toxicol 2017; 72:191-200. [DOI: 10.1016/j.reprotox.2017.05.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 04/20/2017] [Accepted: 05/04/2017] [Indexed: 12/19/2022]
|
25
|
Wang Q, Dong X, Wang Y, Liu M, Sun A, Li N, lin Y, Geng Z, Jin Y, Li X. Adolescent escitalopram prevents the effects of maternal separation on depression‐ and anxiety‐like behaviours and regulates the levels of inflammatory cytokines in adult male mice. Int J Dev Neurosci 2017; 62:37-45. [PMID: 28778811 DOI: 10.1016/j.ijdevneu.2017.07.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 07/25/2017] [Accepted: 07/31/2017] [Indexed: 01/26/2023] Open
Affiliation(s)
- Qi Wang
- Department of PsychiatryThe First Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Xiaomei Dong
- Department of PsychiatryThe First Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Yan Wang
- Department of PsychiatryThe First Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Mengxi Liu
- Department of PsychiatryThe First Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Anji Sun
- Department of PsychiatryThe First Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Nannan Li
- Department of PsychiatryThe First Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Yiwei lin
- Department of PsychiatryThe First Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Zhongli Geng
- Department of PsychiatryThe First Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Ye Jin
- Department of PsychiatryThe First Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Xiaobai Li
- Department of PsychiatryThe First Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| |
Collapse
|
26
|
Zalachoras I, Verhoeve SL, Toonen LJ, van Weert LTCM, van Vlodrop AM, Mol IM, Meelis W, de Kloet ER, Meijer OC. Isoform switching of steroid receptor co-activator-1 attenuates glucocorticoid-induced anxiogenic amygdala CRH expression. Mol Psychiatry 2016; 21:1733-1739. [PMID: 26976039 DOI: 10.1038/mp.2016.16] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 12/01/2015] [Accepted: 01/26/2016] [Indexed: 01/08/2023]
Abstract
Maladaptive glucocorticoid effects contribute to stress-related psychopathology. The glucocorticoid receptor (GR) that mediates many of these effects uses multiple signaling pathways. We have tested the hypothesis that manipulation of downstream factors ('coregulators') can abrogate potentially maladaptive GR-mediated effects on fear-motivated behavior that are linked to corticotropin releasing hormone (CRH). For this purpose the expression ratio of two splice variants of steroid receptor coactivator-1 (SRC-1) was altered via antisense-mediated 'exon-skipping' in the central amygdala of the mouse brain. We observed that a change in splicing towards the repressive isoform SRC-1a strongly reduced glucocorticoid-induced responsiveness of Crh mRNA expression and increased methylation of the Crh promoter. The transcriptional GR target gene Fkbp5 remained responsive to glucocorticoids, indicating gene specificity of the effect. The shift of the SRC-1 splice variants altered glucocorticoid-dependent exploratory behavior and attenuated consolidation of contextual fear memory. In conclusion, our findings demonstrate that manipulation of GR signaling pathways related to the Crh gene can selectively diminish potentially maladaptive effects of glucocorticoids.
Collapse
Affiliation(s)
- I Zalachoras
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - S L Verhoeve
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - L J Toonen
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - L T C M van Weert
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - A M van Vlodrop
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - I M Mol
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - W Meelis
- Department of Medical Pharmacology, Leiden Academic Center for Drug Research, Leiden, The Netherlands
| | - E R de Kloet
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Department of Medical Pharmacology, Leiden Academic Center for Drug Research, Leiden, The Netherlands
| | - O C Meijer
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Institute for Brain and Cognition, Leiden, The Netherlands
| |
Collapse
|
27
|
Pałasz A, Suszka-Świtek A, Filipczyk Ł, Bogus K, Rojczyk E, Worthington J, Krzystanek M, Wiaderkiewicz R. Escitalopram affects spexin expression in the rat hypothalamus, hippocampus and striatum. Pharmacol Rep 2016; 68:1326-1331. [PMID: 27710862 DOI: 10.1016/j.pharep.2016.09.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 08/13/2016] [Accepted: 09/01/2016] [Indexed: 11/28/2022]
Abstract
BACKGROUND Spexin (SPX) is a recently discovered neuropeptide that exhibits a large spectrum of central and peripheral regulatory activity, especially when considered as a potent anorexigenic factor. It has already been proven that antidepressants, including selective serotonin reuptake inhibitors (SSRI), can modulate peptidergic signaling in various brain structures. Despite these findings, there is so far no information regarding the influence of treatment with the SSRI antidepressant escitalopram on brain SPX expression. METHODS In this current study we measured SPX mRNA and protein expression in the selected brain structures (hypothalamus, hippocampus and striatum) of rats chronically treated with a 10mg/kg dose of escitalopram using quantitative Real-Time PCR and immunohistochemistry. RESULTS Strikingly, long-term (4 week) drug treatment led to the downregulation of SPX expression in the rat hypothalamus. This supports the hypothesis that SPX may be involved in the hypothalamic serotonin-dependent actions of SSRI antidepressants and possibly also in the central mechanism of body mass increase. Conversely, SPX expression increased in the hippocampus and striatum. CONCLUSIONS This is the first report of the effects of a neuropsychiatric medication on SPX expression in animal brain. Our findings shed a new light on the pharmacology of antidepressants and may contribute to a better understanding of the alternative mechanisms responsible for antidepressant action.
Collapse
Affiliation(s)
- Artur Pałasz
- Department of Histology, School of Medicine in Katowice, Medical University of Silesia, Medyków 18, Katowice 40-752, Poland.
| | - Aleksandra Suszka-Świtek
- Department of Histology, School of Medicine in Katowice, Medical University of Silesia, Medyków 18, Katowice 40-752, Poland
| | - Łukasz Filipczyk
- Department of Histology, School of Medicine in Katowice, Medical University of Silesia, Medyków 18, Katowice 40-752, Poland
| | - Katarzyna Bogus
- Department of Histology, School of Medicine in Katowice, Medical University of Silesia, Medyków 18, Katowice 40-752, Poland
| | - Ewa Rojczyk
- Department of Histology, School of Medicine in Katowice, Medical University of Silesia, Medyków 18, Katowice 40-752, Poland
| | - John Worthington
- Manchester Immunology Group, Faculty of Life Sciences, University of Manchester, Greater Manchester, M13 9PT, UK; Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, LA1 4YQ, UK
| | - Marek Krzystanek
- Department and Clinic of Psychiatric Rehabilitation, School of Medicine in Katowice, Medical University of Silesia, Ziolowa 45/47, Katowice 40-635, Poland
| | - Ryszard Wiaderkiewicz
- Department of Histology, School of Medicine in Katowice, Medical University of Silesia, Medyków 18, Katowice 40-752, Poland
| |
Collapse
|
28
|
Barra de la Tremblaye P, Plamondon H. Alterations in the corticotropin-releasing hormone (CRH) neurocircuitry: Insights into post stroke functional impairments. Front Neuroendocrinol 2016; 42:53-75. [PMID: 27455847 DOI: 10.1016/j.yfrne.2016.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/04/2016] [Accepted: 07/06/2016] [Indexed: 10/21/2022]
Abstract
Although it is well accepted that changes in the regulation of the hypothalamic-pituitary adrenal (HPA) axis may increase susceptibility to affective disorders in the general population, this link has been less examined in stroke patients. Yet, the bidirectional association between depression and cardiovascular disease is strong, and stress increases vulnerability to stroke. Corticotropin-releasing hormone (CRH) is the central stress hormone of the HPA axis pathway and acts by binding to CRH receptors (CRHR) 1 and 2, which are located in several stress-related brain regions. Evidence from clinical and animal studies suggests a role for CRH in the neurobiological basis of depression and ischemic brain injury. Given its importance in the regulation of the neuroendocrine, autonomic, and behavioral correlates of adaptation and maladaptation to stress, CRH is likely associated in the pathophysiology of post stroke emotional impairments. The goals of this review article are to examine the clinical and experimental data describing (1) that CRH regulates the molecular signaling brain circuit underlying anxiety- and depression-like behaviors, (2) the influence of CRH and other stress markers in the pathophysiology of post stroke emotional and cognitive impairments, and (3) context and site specific interactions of CRH and BDNF as a basis for the development of novel therapeutic targets. This review addresses how the production and release of the neuropeptide CRH within the various regions of the mesocorticolimbic system influences emotional and cognitive behaviors with a look into its role in psychiatric disorders post stroke.
Collapse
Affiliation(s)
- P Barra de la Tremblaye
- School of Psychology, Behavioral Neuroscience Program, University of Ottawa, 136 Jean-Jacques Lussier, Vanier Building, Ottawa, Ontario K1N 6N5, Canada
| | - H Plamondon
- School of Psychology, Behavioral Neuroscience Program, University of Ottawa, 136 Jean-Jacques Lussier, Vanier Building, Ottawa, Ontario K1N 6N5, Canada.
| |
Collapse
|
29
|
Tseilikman VE, Sinitskii AI, Tseilikman OB, Deev RV, Lapshin MS, Kozochkin DA. Glucocorticoid-Related Regulation of LPO in Brain Cortex during Anxiogenic Stress. Bull Exp Biol Med 2015; 159:729-31. [DOI: 10.1007/s10517-015-3060-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Indexed: 12/01/2022]
|
30
|
Mokoena ML, Harvey BH, Viljoen F, Ellis SM, Brink CB. Ozone exposure of Flinders Sensitive Line rats is a rodent translational model of neurobiological oxidative stress with relevance for depression and antidepressant response. Psychopharmacology (Berl) 2015; 232:2921-38. [PMID: 25877744 DOI: 10.1007/s00213-015-3928-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 03/24/2015] [Indexed: 12/18/2022]
Abstract
RATIONALE Major depression has been associated with higher levels of air pollution that in turn leads to neurodegeneration via increased oxidative stress. There is a need for suitable translational animal models to study the role of oxidative stress in depression and antidepressant action. OBJECTIVE Considering the gene X environment hypothesis of depression, the present study investigated the effect of chronic ozone inhalation on depression and anxiety-related behavior, cognition, and brain markers of oxidative stress in the Flinders Sensitive Line (FSL) rat. In addition, response to the antioxidant melatonin, and the antidepressants desipramine or escitalopram, was assessed. METHODS Rats were exposed to ozone (0.0 or 0.3 parts per million (ppm)) per inhalation for 4 h daily for a period of 15 days, while simultaneously receiving saline or the above-mentioned drugs. RESULTS The data indicate that chronic ozone inhalation induced memory impairment, anxiety and depression-like effects, reduced cortical and hippocampal superoxide dismutase and catalase activity, and compromised central monoamine levels similar to that noted in depression. Moreover, the behavioral and neurochemical effects of melatonin, desipramine, and escitalopram were mostly attenuated in the presence of ozone. CONCLUSION Thus, genetically susceptible individuals exposed to high levels of oxidative stress are at higher risk of developing mood and/or an anxiety disorders, showing greater redox imbalance and altered behavior. These animals are also more resistant to contemporary antidepressant treatment. The presented model provides robust face, construct, and predictive validity, suitable for studying neuronal oxidative stress in depression, antidepressant action and mechanisms to prevent neuronal oxidative stress.
Collapse
Affiliation(s)
- Mmalebuso L Mokoena
- Division of Pharmacology, School of Pharmacy, North-West University, Potchefstroom, 2520, South Africa
| | | | | | | | | |
Collapse
|
31
|
Ho PS, Yeh YW, Huang SY, Liang CS. A shift toward T helper 2 responses and an increase in modulators of innate immunity in depressed patients treated with escitalopram. Psychoneuroendocrinology 2015; 53:246-55. [PMID: 25647343 DOI: 10.1016/j.psyneuen.2015.01.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 01/08/2015] [Accepted: 01/09/2015] [Indexed: 10/24/2022]
Abstract
Depression is hypothesized to involve inflammatory processes, and identifying the key cytokines targeted by antidepressant drugs is critical for tailoring treatment to specific cases. However, investigating a limited number of cytokines at one time cannot provide a broad picture of antidepressant-associated immunomodulation. Cytokines act in a network where one could demonstrate pleiotropism, redundancy, synergy, and antagonism with other cytokine functions. This study was aimed at determining whether escitalopram functions as an anti-inflammatory agent and, if so, how it influences cytokine networks. A total of 24 healthy controls and 26 patients with clinical depression requiring inpatient treatment were recruited. A multiplex assay, an efficient tool to simultaneously measure 27 cytokines, was applied in patients with depression before and after 4-week escitalopram treatment. Healthy controls did not take escitalopram and completed cytokine analyses once. We demonstrated that escitalopram increased the levels of interleukin (IL)-1 receptor antagonist and IL-2. Moreover, escitalopram contributed to a shift toward T helper 2 responses and an increase in modulators of innate immunity, leading to a decrease of immune system activation, both innate and adaptive. We suggest that escitalopram modulates the balance of IL-1 and IL-1 receptor antagonist and improves the function and number of T regulatory cells. However, diverse conclusions could be drawn if only a few cytokines were assessed or different significance levels were used. Further studies should investigate a wide range of cytokines in a reliable and valid way, which is key to disentangling the effects of different antidepressants on inflammatory processes.
Collapse
Affiliation(s)
- Pei-Shen Ho
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Yi-Wei Yeh
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC; Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - San-Yuan Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC; Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chih-Sung Liang
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC.
| |
Collapse
|
32
|
Li XF, Hu MH, Li SY, Geach C, Hikima A, Rose S, Greenwood MP, Greenwood M, Murphy D, Poston L, Lightman SL, O'Byrne KT. Overexpression of corticotropin releasing factor in the central nucleus of the amygdala advances puberty and disrupts reproductive cycles in female rats. Endocrinology 2014; 155:3934-44. [PMID: 25051447 DOI: 10.1210/en.2014-1339] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Prolonged exposure to environmental stress activates the hypothalamic-pituitary-adrenal (HPA) axis and generally disrupts the hypothalamic-pituitary-gonadal axis. Because CRF expression in the central nucleus of the amygdala (CeA) is a key modulator in adaptation to chronic stress, and central administration of CRF inhibits the hypothalamic GnRH pulse generator, we tested the hypothesis that overexpression of CRF in the CeA of female rats alters anxiety behavior, dysregulates the HPA axis response to stress, changes pubertal timing, and disrupts reproduction. We used a lentiviral vector to increase CRF expression site specifically in the CeA of preweaning (postnatal day 12) female rats. Overexpression of CRF in the CeA increased anxiety-like behavior in peripubertal rats shown by a reduction in time spent in the open arms of the elevated plus maze and a decrease in social interaction. Paradoxically, puberty onset was advanced but followed by irregular estrous cyclicity and an absence of spontaneous preovulatory LH surges associated with proestrous vaginal cytology in rats overexpressing CRF. Despite the absence of change in basal corticosterone secretion or induced by stress (lipopolysaccharide or restraint), overexpression of CRF in the CeA significantly decreased lipopolysaccharide, but not restraint, stress-induced suppression of pulsatile LH secretion in postpubertal ovariectomized rats, indicating a differential stress responsivity of the GnRH pulse generator to immunological stress and a potential adaptation of the HPA axis to chronic activation of amygdaloid CRF. These data suggest that the expression profile of this key limbic brain CRF system might contribute to the complex neural mechanisms underlying the increasing incidence of early onset of puberty on the one hand and infertility on the other attributed to chronic stress in modern human society.
Collapse
Affiliation(s)
- X F Li
- Division of Women's Health (X.F.L., M.H.H., S.Y.L., C.G., L.P., K.T.O.) and Neurodegenerative Disease Research Group (A.H., S.R.), School of Medicine, King's College London, Guy's Campus, London SE1 1UL, United Kingdom; and Henry Wellcome Laboratory for Integrative Neuroscience and Endocrinology (M.P.G., M.G., D.M., S.L.L.), University of Bristol, Bristol BS13NY, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Bourke CH, Glasper ER, Neigh GN. SSRI or CRF antagonism partially ameliorate depressive-like behavior after adolescent social defeat. Behav Brain Res 2014; 270:295-9. [PMID: 24867331 DOI: 10.1016/j.bbr.2014.05.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 05/13/2014] [Accepted: 05/16/2014] [Indexed: 10/25/2022]
Abstract
Depression and anxiety during adolescence are complex disorders due to persistent effects on physiology and behavior. Selective-serotonin reuptake inhibitors (SSRI) are currently the most widely used pharmacological intervention for depression. Corticotropin-releasing factor one (CRF1) receptor antagonists represent a novel class of compounds that may have efficacy for depressive and anxiety disorders. This study used an animal model of chronic adolescent stress to determine the efficacy of the SSRI fluoxetine, and a novel CRF1 receptor antagonist, GSK876008, on prevention of the behavioral effects of chronic adolescent stress. Male rats were exposed to chronic social defeat stress, fluoxetine, and/or GSK876008 from postnatal day 28-50. Chronic stress-induced depressive-like behaviors were partially attenuated by either concurrent fluoxetine or GSK876008. Fluoxetine blunted body mass gain in the adolescents exposed to chronic stress. The collective data demonstrate similar efficacy between a SSRI and a CRF1 receptor antagonist in the attenuation of stress-induced anhedonia but fewer side effects were observed in those rats treated with the CRF1 receptor antagonist. These data suggest that CRF1 receptor antagonists may be a viable alternative for treatment of depressive behaviors in adolescents.
Collapse
Affiliation(s)
- Chase H Bourke
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Erica R Glasper
- Department of Psychology, University of Maryland, College Park, MD, USA
| | - Gretchen N Neigh
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA; Department of Physiology, Emory University, Atlanta, GA, USA.
| |
Collapse
|
34
|
Kormos V, Gaszner B. Role of neuropeptides in anxiety, stress, and depression: from animals to humans. Neuropeptides 2013; 47:401-19. [PMID: 24210138 DOI: 10.1016/j.npep.2013.10.014] [Citation(s) in RCA: 235] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 10/07/2013] [Accepted: 10/09/2013] [Indexed: 12/11/2022]
Abstract
Major depression, with its strikingly high prevalence, is the most common cause of disability in communities of Western type, according to data of the World Health Organization. Stress-related mood disorders, besides their deleterious effects on the patient itself, also challenge the healthcare systems with their great social and economic impact. Our knowledge on the neurobiology of these conditions is less than sufficient as exemplified by the high proportion of patients who do not respond to currently available medications targeting monoaminergic systems. The search for new therapeutical strategies became therefore a "hot topic" in neuroscience, and there is a large body of evidence suggesting that brain neuropeptides not only participate is stress physiology, but they may also have clinical relevance. Based on data obtained in animal studies, neuropeptides and their receptors might be targeted by new candidate neuropharmacons with the hope that they will become important and effective tools in the management of stress related mood disorders. In this review, we attempt to summarize the latest evidence obtained using animal models for mood disorders, genetically modified rodent models for anxiety and depression, and we will pay some attention to previously published clinical data on corticotropin releasing factor, urocortin 1, urocortin 2, urocortin 3, arginine-vasopressin, neuropeptide Y, pituitary adenylate-cyclase activating polypeptide, neuropeptide S, oxytocin, substance P and galanin fields of stress research.
Collapse
Affiliation(s)
- Viktória Kormos
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Pécs, Szigeti u. 12, H-7624 Pécs, Hungary; Molecular Pharmacology Research Group, János Szentágothai Research Center, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary; Department of Anatomy, Faculty of Medicine, University of Pécs, Szigeti u. 12, H-7624 Pécs, Hungary
| | | |
Collapse
|
35
|
Abstract
BACKGROUND Mood disorders are expressed in many heterogeneous forms, varying from anxiety to severe major clinical depression. The disorders are expressed in individual variety through manifestations governed by co-morbidities, symptom frequency, severity, and duration, and the effects of genes on phenotypes. The underlying etiologies of mood disorders consist of complex interactive operations of genetic and environmental factors. The notion of endophenotypes, which encompasses the markers of several underlying liabilities to the disorders, may facilitate efforts to detect and define, through staging, the genetic risks inherent to the extreme complexity of disease state. AIMS This review evaluates the role of genetic biomarkers in assisting clinical diagnosis, identification of risk factors, and treatment of mood disorders. METHODS Through a systematic assessment of studies investigating the epigenetic basis for mood disorders, the present review examines the interaction of genes and environment underlying the pathophysiology of these disorders. RESULTS The majority of research findings suggest that the notion of endophenotypes, which encompasses the markers of several underlying liabilities to the disorders, may facilitate efforts to detect and define, through staging, the genetic risks inherent to the extreme complexity of the disease states. Several strategies under development and refinement show the propensity for derivation of essential elements in the etiopathogenesis of the disorders affecting drug-efficacy, drug metabolism, and drug adverse effects, e.g., with regard to selective serotonin reuptake inhibitors. These include: transporter gene expression and genes encoding receptor systems, hypothalamic-pituitary-adrenal axis factors, neurotrophic factors, and inflammatory factors affecting neuroimmune function. Nevertheless, procedural considerations of pharmacogenetics presume the parallel investment of policies and regulations to withstand eventual attempts at misuse, thereby ensuring patient integrity. CONCLUSIONS Identification of genetic biomarkers facilitates choice of treatment, prediction of response, and prognosis of outcome over a wide spectrum of symptoms associated with affective states, thereby optimizing clinical practice procedures. Epigenetic regulation of primary brain signaling, e.g., serotonin and hypothalamic-pituitary-adrenal function, and factors governing their metabolism are necessary considerations. The participation of neurotrophic factors remains indispensable for neurogenesis, survival, and functional maintenance of brain systems.
Collapse
Affiliation(s)
- T Archer
- Department of Psychology, University of Gothenburg, Box 500, SE-40530 Gothenburg, Sweden
| | | | | | | |
Collapse
|