1
|
Zhao R, Shi H, Wang Y, Jiang T, Xu Y. Allele-specific methylation of SSTR4 associated with aging and cognitive functions in patients with schizophrenia. PLoS One 2025; 20:e0303038. [PMID: 39908289 PMCID: PMC11798447 DOI: 10.1371/journal.pone.0303038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/18/2024] [Indexed: 02/07/2025] Open
Abstract
The co-occurrence of alcohol use disorder (AUD) and schizophrenia is prevalent, with a rate of 33.7%. Previous research has suggested a genetic and epigenetic overlap between these two disorders. SSTR4, a member of the somatostatin receptor family, is implicated in various neurological and psychiatric conditions, including cognitive function, AUD, and schizophrenia. However, the role of genetic-epigenetic interactions involving SSTR4 in patients with schizophrenia remains unexplored. In this study, we conducted an integration of publicly available datasets and identified allele-specific methylation patterns in SSTR4. Additionally, we pinpointed several genetic variants (rs17691954, rs11464356, rs3109190, and rs145879288) that influence the pace of aging and cognitive functions (rs705935) through their quantitative trait loci effects on CpG sites within SSTR4.
Collapse
Affiliation(s)
- Rongrong Zhao
- The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Huihui Shi
- The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Yanqiu Wang
- The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Tao Jiang
- The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Yahui Xu
- The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
2
|
Zhang B, Xue L, Wu ZB. Structure and Function of Somatostatin and Its Receptors in Endocrinology. Endocr Rev 2025; 46:26-42. [PMID: 39116368 DOI: 10.1210/endrev/bnae022] [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: 12/27/2023] [Revised: 07/16/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024]
Abstract
Somatostatin analogs, such as octreotide, lanreotide, and pasireotide, which function as somatostatin receptor ligands (SRLs), are the main drugs used for the treatment of acromegaly. These ligands are also used as important molecules for radiation therapy and imaging of neuroendocrine tumors. Somatostatin receptors (SSTRs) are canonical G protein-coupled proteins that play a role in metabolism, growth, and pathological conditions such as hormone disorders, neurological diseases, and cancers. Cryogenic electron microscopy combined with the protein structure prediction platform AlphaFold has been used to determine the 3-dimensional structures of many proteins. Recently, several groups published a series of papers illustrating the 3-dimensional structure of SSTR2, including that of the inactive/activated SSTR2-G protein complex bound to different ligands. The results revealed the residues that contribute to the ligand binding pocket and demonstrated that Trp8-Lys9 (the W-K motif) in somatostatin analogs is the key motif in stabilizing the bottom part of the binding pocket. In this review, we discuss the recent findings related to the structural analysis of SSTRs and SRLs, the relationships between the structural data and clinical findings, and the future development of novel structure-based therapies.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Li Xue
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhe Bao Wu
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325005, China
| |
Collapse
|
3
|
Valente R, Cordeiro M, Pinto B, Machado A, Alves F, Sousa-Pinto I, Ruivo R, Castro LFC. Alterations of pleiotropic neuropeptide-receptor gene couples in Cetacea. BMC Biol 2024; 22:186. [PMID: 39218857 PMCID: PMC11367936 DOI: 10.1186/s12915-024-01984-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Habitat transitions have considerable consequences in organism homeostasis, as they require the adjustment of several concurrent physiological compartments to maintain stability and adapt to a changing environment. Within the range of molecules with a crucial role in the regulation of different physiological processes, neuropeptides are key agents. Here, we examined the coding status of several neuropeptides and their receptors with pleiotropic activity in Cetacea. RESULTS Analysis of 202 mammalian genomes, including 41 species of Cetacea, exposed an intricate mutational landscape compatible with gene sequence modification and loss. Specifically for Cetacea, in the 12 genes analysed we have determined patterns of loss ranging from species-specific disruptive mutations (e.g. neuropeptide FF-amide peptide precursor; NPFF) to complete erosion of the gene across the cetacean stem lineage (e.g. somatostatin receptor 4; SSTR4). CONCLUSIONS Impairment of some of these neuromodulators may have contributed to the unique energetic metabolism, circadian rhythmicity and diving response displayed by this group of iconic mammals.
Collapse
Affiliation(s)
- Raul Valente
- CIMAR/CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, 4450-208, Matosinhos, S/N, Portugal
- FCUP - Department of Biology, Faculty of Sciences, University of Porto (U. Porto), Rua Do Campo Alegre, Porto, Portugal
| | - Miguel Cordeiro
- CIMAR/CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, 4450-208, Matosinhos, S/N, Portugal
| | - Bernardo Pinto
- CIMAR/CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, 4450-208, Matosinhos, S/N, Portugal
- FCUP - Department of Biology, Faculty of Sciences, University of Porto (U. Porto), Rua Do Campo Alegre, Porto, Portugal
| | - André Machado
- CIMAR/CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, 4450-208, Matosinhos, S/N, Portugal
- FCUP - Department of Biology, Faculty of Sciences, University of Porto (U. Porto), Rua Do Campo Alegre, Porto, Portugal
| | - Filipe Alves
- MARE - Marine and Environmental Sciences Centre, Funchal, Madeira, Portugal
- ARNET - Aquatic Research Network, ARDITI, Funchal, Madeira, Portugal
| | - Isabel Sousa-Pinto
- CIMAR/CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, 4450-208, Matosinhos, S/N, Portugal
- FCUP - Department of Biology, Faculty of Sciences, University of Porto (U. Porto), Rua Do Campo Alegre, Porto, Portugal
| | - Raquel Ruivo
- CIMAR/CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, 4450-208, Matosinhos, S/N, Portugal.
| | - L Filipe C Castro
- CIMAR/CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, 4450-208, Matosinhos, S/N, Portugal.
- FCUP - Department of Biology, Faculty of Sciences, University of Porto (U. Porto), Rua Do Campo Alegre, Porto, Portugal.
| |
Collapse
|
4
|
Periferakis A, Tsigas G, Periferakis AT, Tone CM, Hemes DA, Periferakis K, Troumpata L, Badarau IA, Scheau C, Caruntu A, Savulescu-Fiedler I, Caruntu C, Scheau AE. Agonists, Antagonists and Receptors of Somatostatin: Pathophysiological and Therapeutical Implications in Neoplasias. Curr Issues Mol Biol 2024; 46:9721-9759. [PMID: 39329930 PMCID: PMC11430067 DOI: 10.3390/cimb46090578] [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: 07/31/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/28/2024] Open
Abstract
Somatostatin is a peptide that plays a variety of roles such as neurotransmitter and endocrine regulator; its actions as a cell regulator in various tissues of the human body are represented mainly by inhibitory effects, and it shows potent activity despite its physiological low concentrations. Somatostatin binds to specific receptors, called somatostatin receptors (SSTRs), which have different tissue distributions and associated signaling pathways. The expression of SSTRs can be altered in various conditions, including tumors; therefore, they can be used as biomarkers for cancer cell susceptibility to certain pharmacological agents and can provide prognostic information regarding disease evolution. Moreover, based on the affinity of somatostatin analogs for the different types of SSTRs, the therapeutic range includes conditions such as tumors, acromegaly, post-prandial hypotension, hyperinsulinism, and many more. On the other hand, a number of somatostatin antagonists may prove useful in certain medical settings, based on their differential affinity for SSTRs. The aim of this review is to present in detail the principal characteristics of all five SSTRs and to provide an overview of the associated therapeutic potential in neoplasias.
Collapse
Affiliation(s)
- Argyrios Periferakis
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Elkyda, Research & Education Centre of Charismatheia, 17675 Athens, Greece
- Akadimia of Ancient Greek and Traditional Chinese Medicine, 16675 Athens, Greece
| | - Georgios Tsigas
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Aristodemos-Theodoros Periferakis
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Elkyda, Research & Education Centre of Charismatheia, 17675 Athens, Greece
| | - Carla Mihaela Tone
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Daria Alexandra Hemes
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Konstantinos Periferakis
- Akadimia of Ancient Greek and Traditional Chinese Medicine, 16675 Athens, Greece
- Pan-Hellenic Organization of Educational Programs, 17236 Athens, Greece
| | - Lamprini Troumpata
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Ioana Anca Badarau
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Radiology and Medical Imaging, "Foisor" Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 030167 Bucharest, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, The "Carol Davila" Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, "Titu Maiorescu" University, 031593 Bucharest, Romania
| | - Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, "Prof. N.C. Paulescu" National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| |
Collapse
|
5
|
Zhao R, Shi H, Wang Y, Zheng S, Xu Y. Methylation of SSTR4 promoter region in multiple mental health disorders. Front Genet 2024; 15:1431769. [PMID: 39055257 PMCID: PMC11269100 DOI: 10.3389/fgene.2024.1431769] [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: 05/12/2024] [Accepted: 06/10/2024] [Indexed: 07/27/2024] Open
Abstract
The existence of a shared genetic basis for mental disorders has long been documented, yet research on whether acquired epigenetic modifications exhibit common alterations across diseases is limited. Previous studies have found that abnormal methylation of cg14631053 at the SSTR4 promoter region mediates the onset of alcohol use disorder. However, whether aberrant methylation of the SSTR4 gene promoter is involved in other mental health disorders remains unclear. In this study, leveraging publicly available data, we identified that changes in methylation of cg14631053 from the SSTR4 promoter region are involved in the development of bipolar disorder and schizophrenia. Furthermore, the direction of methylation changes in the SSTR4 promoter region is disease-specific: hypomethylation is associated with the onset of bipolar disorder and schizophrenia, rather than major depressive disorder. Methylation levels of cg14631053 correlate with chronological age, a correlation that can be disrupted in patients with mental health disorders including schizophrenia and bipolar disorder. In conclusion, SSTR4 promoter methylation may serve as a marker for identifying bipolar disorder and schizophrenia, providing insights into a transdiagnostic mechanism for precision medicine in the future.
Collapse
Affiliation(s)
- Rongrong Zhao
- The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Huihui Shi
- The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Yanqiu Wang
- The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Shuaiyu Zheng
- The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Yahui Xu
- The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
6
|
Abe Y, Murase T, Mitsuma M, Shinba Y, Yamashita H, Ikematsu K. Dynamics of somatostatin 4 receptor expression during chronic-stress loading and its potential as a chronic-stress marker. Sci Rep 2024; 14:10045. [PMID: 38698013 PMCID: PMC11066077 DOI: 10.1038/s41598-024-58621-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 04/01/2024] [Indexed: 05/05/2024] Open
Abstract
Chronic stress has been implicated in mental illnesses and depressive behaviors. Somatostatin 4 receptor (SSTR4) has been shown to mediate anxiolytic and depression-like effects. Here, we aimed to explore the potential of SSTR4 as a diagnostic marker for chronic stress in mice. The mice were divided into single stress, chronic restraint stress, and control groups, and Sstr4 mRNA expression in the pituitary, lungs, and thymus, its protein expression in the thymus, were analyzed. Compared to controls, Sstr4 mRNA expression decreased significantly in the pituitary gland of the chronic and single-stress groups (P = 0.0181 and 0.0022, respectively) and lungs of the single-stress group (P = 0.0124), whereas it significantly increased in the thymus of the chronic-stress group (P = 0.0313). Thymic SSTR4 expression did not decrease significantly in stress groups compared to that in the control group (P = 0.0963). These results suggest that SSTR4 expression fluctuates in response to stress. Furthermore, Sstr4 mRNA expression dynamics in each organ differed based on single or chronic restraint stress-loading periods. In conclusion, this study suggests that investigating SSTR4 expression in each organ could allow for its use as a stress marker to estimate the stress-loading period and aid in diagnosing chronic stress.
Collapse
Affiliation(s)
- Yuki Abe
- Division of Forensic Pathology and Science, Department of Medical and Dental Sciences, Graduate School of Biomedical Sciences, School of Medicine, Nagasaki University, 1-12-4, Sakamoto, Nagasaki, 852-8523, Japan
| | - Takehiko Murase
- Division of Forensic Pathology and Science, Department of Medical and Dental Sciences, Graduate School of Biomedical Sciences, School of Medicine, Nagasaki University, 1-12-4, Sakamoto, Nagasaki, 852-8523, Japan.
- Department of Forensic Medicine, Faculty of Medicine, Kagawa University, 1750-1, Miki, Kita, Kagawa, 761-0793, Japan.
| | - Masahide Mitsuma
- Division of Forensic Pathology and Science, Department of Medical and Dental Sciences, Graduate School of Biomedical Sciences, School of Medicine, Nagasaki University, 1-12-4, Sakamoto, Nagasaki, 852-8523, Japan
| | - Yoriko Shinba
- Division of Forensic Pathology and Science, Department of Medical and Dental Sciences, Graduate School of Biomedical Sciences, School of Medicine, Nagasaki University, 1-12-4, Sakamoto, Nagasaki, 852-8523, Japan
| | - Hiromi Yamashita
- Division of Forensic Dental Science, Department of Medical and Dental Sciences, Graduate School of Biomedical Sciences, School of Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Kazuya Ikematsu
- Division of Forensic Pathology and Science, Department of Medical and Dental Sciences, Graduate School of Biomedical Sciences, School of Medicine, Nagasaki University, 1-12-4, Sakamoto, Nagasaki, 852-8523, Japan
| |
Collapse
|
7
|
Wang R, Hu X, Liu S, Wang J, Xiong F, Zhang X, Ye W, Wang H. Kaempferol-3-O-sophoroside (PCS-1) contributes to modulation of depressive-like behaviour in C57BL/6J mice by activating AMPK. Br J Pharmacol 2024; 181:1182-1202. [PMID: 37949672 DOI: 10.1111/bph.16283] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 10/18/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND AND PURPOSE Kaempferol-3-O-sophoroside (PCS-1) is the main component in Crocus sativus (Saffron), a herb with mood-enhancing properties. AMP-activated protein kinase (AMPK) is a potential therapeutic target for depression. This study explores the antidepressive-like properties of PCS-1 and its AMPK activation to confirm AMPK as a target for antidepression. EXPERIMENTAL APPROACH Corticosterone (CORT)-induced PC12 cell injury served as an in vitro model to evaluate the neuroprotective effect of PCS-1. Neuro-2a cells and primary neurons were utilized to evaluate the synaptogenesis role of PCS-1. CORT-induced mouse depression model and chronic unpredictable mild stress (CUMS) model were used to assess the antidepressive-like properties of PCS-1 through behavioural tests, magnetic resonance imaging, and biochemical index measurements. Western blot and immunofluorescence assays were used to study the mechanisms of PCS-1. Cellular thermal shift assay was used to confirm the binding target. KEY RESULTS PCS-1 (12.5-50 μM) ameliorated CORT-induced PC12 cell damage, oxidative stress and inflammation. PCS-1 alone promoted an increase in synapses in Neuro-2a cells and primary neurons. Oral administration of PCS-1 (10 and 20 mg·kg-1 ) ameliorated weight loss, dyskinesia, and hippocampal volume reduction induced by CORT and CUMS. PCS-1 bound to AMPK to improve the expression of brain-derived neurotrophic factor (BDNF) and induce autophagy. CONCLUSION AND IMPLICATIONS PCS-1 binds to AMPK to promote BDNF production and autophagy enhancement, ultimately achieving antidepressant effects. This study provides support for the clinical application of saffron petals and provides further evidence for AMPK as a potential target for antidepression.
Collapse
Affiliation(s)
- Rong Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Xiaolong Hu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Shumeng Liu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Jingjin Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Fei Xiong
- State Key Laboratory of Bioelectronics, Jiangsu Laboratory for Biomaterials and Devices, Southeast University, Nanjing, People's Republic of China
| | - Xiaoqi Zhang
- Institute of Traditional Chinese Medicine & Natural Products, Jinan University, Guangzhou, People's Republic of China
| | - Wencai Ye
- Institute of Traditional Chinese Medicine & Natural Products, Jinan University, Guangzhou, People's Republic of China
| | - Hao Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
8
|
Borbély É, Kecskés A, Kun J, Kepe E, Fülöp B, Kovács-Rozmer K, Scheich B, Renner É, Palkovits M, Helyes Z. Hemokinin-1 is a mediator of chronic restraint stress-induced pain. Sci Rep 2023; 13:20030. [PMID: 37973885 PMCID: PMC10654722 DOI: 10.1038/s41598-023-46402-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 10/31/2023] [Indexed: 11/19/2023] Open
Abstract
The Tac4 gene-derived hemokinin-1 (HK-1) binds to the NK1 receptor, similarly to Substance P, and plays a role in acute stress reactions and pain transmission in mice. Here we investigated Tac4 mRNA expression in stress and pain-related regions and its involvement in chronic restraint stress-evoked behavioral changes and pain using Tac4 gene-deleted (Tac4-/-) mice compared to C57Bl/6 wildtypes (WT). Tac4 mRNA was detected by in situ hybridization RNAscope technique. Touch sensitivity was assessed by esthesiometry, cold tolerance by paw withdrawal latency from 0°C water. Anxiety was evaluated in the light-dark box (LDB) and open field test (OFT), depression-like behavior in the tail suspension test (TST). Adrenal and thymus weights were measured at the end of the experiment. We found abundant Tac4 expression in the hypothalamic-pituitary-adrenal axis, but Tac4 mRNA was also detected in the hippocampus, amygdala, somatosensory and piriform cortices in mice, and in the frontal regions and the amygdala in humans. In Tac4-/- mice of both sexes, stress-induced mechanical, but not cold hyperalgesia was significantly decreased compared to WTs. Stress-induced behavioral alterations were mild or absent in male WT animals, while significant changes of these parameters could be detected in females. Thymus weight decrease can be observed in both sexes. Higher baseline anxiety and depression-like behaviors were detected in male but not in female HK-1-deficient mice, highlighting the importance of investigating both sexes in preclinical studies. We provided the first evidence for the potent nociceptive and stress regulating effects of HK-1 in chronic restraint stress paradigm. Identification of its targets might open new perspectives for therapy of stress-induced pain.
Collapse
Affiliation(s)
- Éva Borbély
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary.
- Centre for Neuroscience, University of Pécs, Pécs, Hungary.
| | - Angéla Kecskés
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - József Kun
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - Eszter Kepe
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Barbara Fülöp
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Katalin Kovács-Rozmer
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- Chronic Pain Research Group, Hungarian Research Network, University of Pécs, Pécs, Hungary
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | - Bálint Scheich
- Department of Pathology and Experimental Cancer Research, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Éva Renner
- Human Brain Tissue Bank, Semmelweis University, Budapest, Hungary
| | - Miklós Palkovits
- Human Brain Tissue Bank, Semmelweis University, Budapest, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, Hungary
- PharmInVivo Ltd, Pécs, Hungary
- Chronic Pain Research Group, Hungarian Research Network, University of Pécs, Pécs, Hungary
- National Laboratory for Drug Research and Development, Magyar Tudósok Krt. 2, Budapest, 1117, Hungary
| |
Collapse
|
9
|
Robinson SL, Thiele TE. Somatostatin signaling modulates binge drinking behavior via the central nucleus of the amygdala. Neuropharmacology 2023; 237:109622. [PMID: 37307896 PMCID: PMC10527233 DOI: 10.1016/j.neuropharm.2023.109622] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 06/02/2023] [Accepted: 06/02/2023] [Indexed: 06/14/2023]
Abstract
Somatostatin (SST) is a neuropeptide widely expressed in the central nervous system with dense expression in limbic regions such as the extended amygdala. It has recently gained attention for playing a role in modulating alcohol use disorders and co-morbid neuropsychiatric disorders. However, the role of SST in the central nucleus of the amygdala (CeA), a key region for neuropeptide regulation of alcohol and anxiety related behaviors, in alcohol consumption has not been assessed. In this work we perform an initial examination of the interaction between the CeA SST system and binge ethanol intake. Binge intake is a dangerous pattern of excessive ethanol consumption associated with health complications and the transition into alcohol dependence. We use the Drinking in the Dark (DID) model of binge intake in C57BL/6J male and female mice to examine: 1) the impact of 3 DID cycles on CeA SST expression; 2) the effect of intra-CeA SST injection on binge-like ethanol consumption; and 3) if the SST receptor 2 or 4 (SST2R or SST4R) mediate any effect on consumption. Our results show binge-like ethanol intake decreases SST expression in the CeA, but not neighboring basolateral amygdala. We further found intra-SST CeA administration reduces binge ethanol intake. This decrease was replicated by the administration of an SST4R agonist. These effects were not sex-dependent. Overall, this work lends further support for SST playing a role in alcohol related behaviors and as a potential therapeutic target.
Collapse
Affiliation(s)
- Stacey L Robinson
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC, 27599, USA; Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Todd E Thiele
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC, 27599, USA; Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
10
|
Ngoc KH, Kecskés A, Kepe E, Nabi L, Keeble J, Borbély É, Helyes Z. Expression of the Transient Receptor Potential Vanilloid 1 ion channel in the supramammillary nucleus and the antidepressant effects of its antagonist AMG9810 in mice. Eur Neuropsychopharmacol 2023; 73:96-107. [PMID: 37156112 DOI: 10.1016/j.euroneuro.2023.04.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/19/2023] [Accepted: 04/24/2023] [Indexed: 05/10/2023]
Abstract
The Transient Receptor Potential Vanilloid 1 (TRPV1) non-selective cation channel predominantly expressed in primary sensory neurons of the dorsal root and trigeminal ganglia mediates pain and neurogenic inflammation. TRPV1 mRNA and immunoreactivity were described in the central nervous system (CNS), but its precise expression pattern and function have not been clarified. Here we investigated Trpv1 mRNA expression in the mouse brain using ultrasensitive RNAScope in situ hybridization. The role of TRPV1 in anxiety, depression-like behaviors and memory functions was investigated by TRPV1-deficient mice and pharmacological antagonism by AMG9810. Trpv1 mRNA is selectively expressed in the supramammillary nucleus (SuM) co-localized with Vglut2 mRNA, but not with tyrosine hydroxylase immunopositivity demonstrating its presence in glutamatergic, but not dopaminergic neurons. TRPV1-deleted mice exhibited significantly reduced anxiety in the Light-Dark box and depression-like behaviors in the Forced Swim Test, but their performance in the Elevated Plus Maze as well as their spontaneous locomotor activity, memory and learning function in the Radial Arm Maze, Y-maze and Novel Object Recognition test were not different from WTs. AMG9810 (intraperitoneal injection 50 mg/kg) induced anti-depressant, but not anxiolytic effects. It is concluded that TRPV1 in the SuM might have functional relevance in mood regulation and TRPV1 antagonism could be a novel perspective for anti-depressant drugs.
Collapse
Affiliation(s)
- Khai Huynh Ngoc
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary; Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - Angéla Kecskés
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - Eszter Kepe
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - Liza Nabi
- King's College London, Institute of Pharmaceutical Science, London, United Kingdom
| | - Julie Keeble
- King's College London, Centre for Human & Applied Physiological Sciences, London, United Kingdom
| | - Éva Borbély
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary; National Laboratory for Drug Research and Development, Budapest, Hungary.
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary; Eötvös Loránd Research Network, Chronic Pain Research Group, University of Pécs, Hungary; National Laboratory for Drug Research and Development, Budapest, Hungary; PharmInVivo Ltd, Pécs, Hungary.
| |
Collapse
|
11
|
Nemes B, László S, Zsidó BZ, Hetényi C, Feher A, Papp F, Varga Z, Szőke É, Sándor Z, Pintér E. Elucidation of the binding mode of organic polysulfides on the human TRPA1 receptor. Front Physiol 2023; 14:1180896. [PMID: 37351262 PMCID: PMC10282659 DOI: 10.3389/fphys.2023.1180896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/22/2023] [Indexed: 06/24/2023] Open
Abstract
Introduction: Previous studies have established that endogenous inorganic polysulfides have significant biological actions activating the Transient Receptor Potential Ankyrin 1 (TRPA1) receptor. Organic polysulfides exert similar effects, but they are much more stable molecules, therefore these compounds are more suitable as drugs. In this study, we aimed to better understand the mechanism of action of organic polysulfides by identification of their binding site on the TRPA1 receptor. Methods: Polysulfides can readily interact with the thiol side chain of the cysteine residues of the protein. To investigate their role in the TRPA1 activation, we replaced several cysteine residues by alanine via site-directed mutagenesis. We searched for TRPA1 mutant variants with decreased or lost activating effect of the polysulfides, but with other functions remaining intact (such as the effects of non-electrophilic agonists and antagonists). The binding properties of the mutant receptors were analyzed by in silico molecular docking. Functional changes were tested by in vitro methods: calcium sensitive fluorescent flow cytometry, whole-cell patch-clamp and radioactive calcium-45 liquid scintillation counting. Results: The cysteines forming the conventional binding site of electrophilic agonists, namely C621, C641 and C665 also bind the organic polysulfides, with the key role of C621. However, only their combined mutation abolished completely the organic polysulfide-induced activation of the receptor. Discussion: Since previous papers provided evidence that organic polysulfides exert analgesic and anti-inflammatory actions in different in vivo animal models, we anticipate that the development of TRPA1-targeted, organic polysulfide-based drugs will be promoted by this identification of the binding site.
Collapse
Affiliation(s)
- Balázs Nemes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - Szabolcs László
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- Department of Inorganic and Analytical Chemistry, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary
| | - Balázs Zoltán Zsidó
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - Csaba Hetényi
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - Adam Feher
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ferenc Papp
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltan Varga
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Éva Szőke
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - Zoltán Sándor
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - Erika Pintér
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
12
|
Zhong J, Li C, Peng L, Pan Y, Yang Y, Guo Q, Zhong T. Repeated neonatal isoflurane exposure facilitated stress-related fear extinction impairment in male mice and was associated with ΔFosB accumulation in the basolateral amygdala and the hippocampal dentate gyrus. Behav Brain Res 2023; 446:114416. [PMID: 37003493 DOI: 10.1016/j.bbr.2023.114416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/15/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023]
Abstract
Volatile anesthetics elicit neurodevelopmental toxicity in rodents and primates and lead to more exaggerated anxiety-like behavior in response to future stress. Anxiety and fear are closely correlated and maladaptive fear-associated learning is regarded as the core mechanism underlying anxiety-related disorders. However, little is known about the interaction between early-life anesthetic exposure and future stress and the accompanying effect on fear-associated learning. In the present study, we evaluated the alterations in fear-associated learning (fear acquisition and extinction) occurring in mice receiving repeated neonatal isoflurane exposure and chronic variable stress (CVS) successively through a series of fear conditioning, fear reinforcing, and fear extinction paradigms. The corticosterone (CORT) response during CVS and the immunohistochemical levels of ΔFosB and c-Fos expression in the basolateral amygdala (BLA) and the hippocampal dentate gyrus (DG) after the extinction retrieval test were also investigated. The results showed that neonatal isoflurane exposure could increase CORT levels following the first diurnal CVS procedure, but not after completion of the whole CVS paradigm. Neonatal isoflurane exposure exerted a repressive effect on fear acquisition, in contrast to that seen with CVS. Neonatal isoflurane exposure and CVS both exerted suppressive effects on fear extinction and there was a significant synergy between them. Furthermore, neonatal isoflurane exposure facilitated CVS-mediated ΔFosB accumulation in the BLA and the hippocampal DG, which may have been responsible for c-Fos expression deficits and fear extinction impairment. Collectively, these findings contribute to the understanding of the interaction between early-life anesthetic exposure and future stress, as well as the accompanying behavioral alterations.
Collapse
Affiliation(s)
- JiaLing Zhong
- Department of Anaesthesiology and Operating Theatre Services, Xiangya Hospital of Central South University, Changsha city, Hunan Province, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha city, Hunan Province, PR China
| | - ChunLin Li
- Department of Anaesthesiology and Operating Theatre Services, Xiangya Hospital of Central South University, Changsha city, Hunan Province, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha city, Hunan Province, PR China
| | - LuoFang Peng
- Department of Anaesthesiology and Operating Theatre Services, Xiangya Hospital of Central South University, Changsha city, Hunan Province, PR China; Teaching and Research Section of Clinical Nursing, Xiangya Hospital of Central South University, Changsha city, Hunan Province, PR China.
| | - Yudan Pan
- Department of Anaesthesiology and Operating Theatre Services, Xiangya Hospital of Central South University, Changsha city, Hunan Province, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha city, Hunan Province, PR China
| | - Yong Yang
- Department of Anaesthesiology and Operating Theatre Services, Xiangya Hospital of Central South University, Changsha city, Hunan Province, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha city, Hunan Province, PR China
| | - QuLian Guo
- Department of Anaesthesiology and Operating Theatre Services, Xiangya Hospital of Central South University, Changsha city, Hunan Province, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha city, Hunan Province, PR China
| | - Tao Zhong
- Department of Anaesthesiology and Operating Theatre Services, Xiangya Hospital of Central South University, Changsha city, Hunan Province, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha city, Hunan Province, PR China.
| |
Collapse
|
13
|
Kraeuter AK. The use of integrated behavioural z-scoring in behavioural neuroscience - A perspective article. J Neurosci Methods 2023; 384:109751. [PMID: 36435327 DOI: 10.1016/j.jneumeth.2022.109751] [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: 09/02/2022] [Revised: 11/16/2022] [Accepted: 11/22/2022] [Indexed: 11/24/2022]
Abstract
Complex pathophysiology in psychiatric disorders results in difficulties interpreting pre-clinical data. Guilloux et al. (2011b), proposed an integrated behavioural z-scoring procedure to improve the predictive validity of animal models by converging evidence similarly used to diagnose mental health conditions in humans. Here, I set out to give a brief review of the current methodology and literature using integrated behavioural z-scoring. Secondly, I will discuss the benefits and downfalls of integrated behavioural z-scoring and its potential future applications. Integrated behavioural z-scoring is a methodology used most frequently within animal models of depression and anxiety. Here, I am suggesting broadening the application of integrated behavioural z-scoring beyond the field of depression and anxiety to a three-step methodology to obtain disease-specific behavioural z-scores (i.e Schizophrenia index, Alzheimer's disease index) to aid translatability and interpretation of data. Lastly, I suggest integrating not only behaviour but also biological variables to create converging psychological and physiological evidence to sustain face and construct validity, while improving predict validity.
Collapse
Affiliation(s)
- Ann-Katrin Kraeuter
- Faculty of Health and Life Sciences, Psychology, Northumbria University, Newcastle upon Tyne, UK.
| |
Collapse
|
14
|
Pintér E, Helyes Z, Szőke É, Bölcskei K, Kecskés A, Pethő G. The triple function of the capsaicin-sensitive sensory neurons: In memoriam János Szolcsányi. Temperature (Austin) 2022; 10:13-34. [PMID: 38059854 PMCID: PMC10177685 DOI: 10.1080/23328940.2022.2147388] [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: 08/31/2022] [Revised: 11/08/2022] [Accepted: 11/08/2022] [Indexed: 11/23/2022] Open
Abstract
This paper is dedicated to the memory of János Szolcsányi (1938-2018), an outstanding Hungarian scientist. Among analgesics that act on pain receptors, he identified capsaicin as a selective lead molecule. He studied the application of capsaicin and revealed several physiological (pain, thermoregulation) and pathophysiological (inflammation, gastric ulcer) mechanisms. He discovered a new neuroregulatory system without sensory efferent reflex and investigated its pharmacology. The authors of this review are his former Ph.D. students who carried out their doctoral work in Szolcsányi's laboratory between 1985 and 2010 and report on the scientific results obtained under his guidance. His research group provided evidence for the triple function of the peptidergic capsaicin-sensitive sensory neurons including classical afferent function, local efferent responses, and remote, hormone-like anti-inflammatory, and antinociceptive actions. They also proposed somatostatin receptor type 4 as a promising drug target for the treatment of pain and inflammation. They revealed that neonatal capsaicin treatment caused no acute neuronal death but instead long-lasting selective ultrastructural and functional changes in B-type sensory neurons, similar to adult treatment. They described that lipid raft disruption diminished the agonist-induced channel opening of the TRPV1, TRPA1, and TRPM8 receptors in native sensory neurons. Szolcsányi's group has developed new devices for noxious heat threshold measurement: an increasing temperature hot plate and water bath. This novel approach proved suitable for assessing the thermal antinociceptive effects of analgesics as well as for analyzing peripheral mechanisms of thermonociception.
Collapse
Affiliation(s)
- Erika Pintér
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary
- National Laboratory for Drug Research and Development, Magyar tudósok krt. 2. H-1117Budapest, Hungary
- Eötvös Lorand Research Network, Chronic Pain Research Group, University of Pécs, H7624, Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary
- National Laboratory for Drug Research and Development, Magyar tudósok krt. 2. H-1117Budapest, Hungary
- Eötvös Lorand Research Network, Chronic Pain Research Group, University of Pécs, H7624, Pécs, Hungary
| | - Éva Szőke
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary
- National Laboratory for Drug Research and Development, Magyar tudósok krt. 2. H-1117Budapest, Hungary
- Eötvös Lorand Research Network, Chronic Pain Research Group, University of Pécs, H7624, Pécs, Hungary
| | - Kata Bölcskei
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary
| | - Angéla Kecskés
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary
| | - Gábor Pethő
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus Str. 2, H-7624 , Pécs, Hungary
| |
Collapse
|
15
|
Neuwirth LS, Verrengia MT, Harikinish-Murrary ZI, Orens JE, Lopez OE. Under or Absent Reporting of Light Stimuli in Testing of Anxiety-Like Behaviors in Rodents: The Need for Standardization. Front Mol Neurosci 2022; 15:912146. [PMID: 36061362 PMCID: PMC9428565 DOI: 10.3389/fnmol.2022.912146] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
Behavioral neuroscience tests such as the Light/Dark Test, the Open Field Test, the Elevated Plus Maze Test, and the Three Chamber Social Interaction Test have become both essential and widely used behavioral tests for transgenic and pre-clinical models for drug screening and testing. However, as fast as the field has evolved and the contemporaneous involvement of technology, little assessment of the literature has been done to ensure that these behavioral neuroscience tests that are crucial to pre-clinical testing have well-controlled ethological motivation by the use of lighting (i.e., Lux). In the present review paper, N = 420 manuscripts were examined from 2015 to 2019 as a sample set (i.e., n = ~20–22 publications per year) and it was found that only a meager n = 50 publications (i.e., 11.9% of the publications sampled) met the criteria for proper anxiogenic and anxiolytic Lux reported. These findings illustrate a serious concern that behavioral neuroscience papers are not being vetted properly at the journal review level and are being released into the literature and public domain making it difficult to assess the quality of the science being reported. This creates a real need for standardizing the use of Lux in all publications on behavioral neuroscience techniques within the field to ensure that contributions are meaningful, avoid unnecessary duplication, and ultimately would serve to create a more efficient process within the pre-clinical screening/testing for drugs that serve as anxiolytic compounds that would prove more useful than what prior decades of work have produced. It is suggested that improving the standardization of the use and reporting of Lux in behavioral neuroscience tests and the standardization of peer-review processes overseeing the proper documentation of these methodological approaches in manuscripts could serve to advance pre-clinical testing for effective anxiolytic drugs. This report serves to highlight this concern and proposes strategies to proactively remedy them as the field moves forward for decades to come.
Collapse
Affiliation(s)
- Lorenz S. Neuwirth
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, United States
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, NY, United States
- *Correspondence: Lorenz S. Neuwirth
| | - Michael T. Verrengia
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, United States
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, NY, United States
| | - Zachary I. Harikinish-Murrary
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, United States
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, NY, United States
| | - Jessica E. Orens
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, United States
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, NY, United States
| | - Oscar E. Lopez
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, United States
- SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury, NY, United States
| |
Collapse
|
16
|
Zhao R, Shi H, Yin J, Sun Z, Xu Y. Promoter Specific Methylation of SSTR4 is Associated With Alcohol Dependence in Han Chinese Males. Front Genet 2022; 13:915513. [PMID: 35754825 PMCID: PMC9218598 DOI: 10.3389/fgene.2022.915513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Alcohol dependence (AD), a disease can be affected by environmental factors with epigenetic modification like DNA methylation changes, is one of the most serious and complex public health problems in China and worldwide. Previous findings from our laboratory using the Illumina Infinium Human Methylation450 BeadChip suggested that methylation at the promoter of SSTR4 was one of the major form of DNA modification in alcohol-dependent populations. To investigate whether DNA methylation levels of the SSTR4 promoter influence alcohol-dependent behaviors, genomic DNA was extracted from the peripheral blood sample of 63 subjects with AD and 65 healthy controls, and pyrosequencing was used to verify the results of BeadChip array. Linear regression was used to analyze the correlation between the methylation levels of SSTR4 promoter and the scores of alcohol dependence scales. Gene expression of SSTR4 in brain tissue was obtained from the Genotype-Tissue Expression (GTEx) project and Human Brain Transcriptome database (HBT). We found the methylation levels of SSTR4 in AD group were significantly lower than healthy controls (two-tailed t-test, t = 14.723, p < 0.001). In addition, only weak to moderate correlations between the methylation levels of the SSTR4 promoter region and scale scores of Alcohol Use Disorders Identification Test (AUDIT), Life Events Scale (LES) and Wheatley Stress Profile (WSS) based on linear regression analyses (AUDIT: R 2 = 0.35, p < 0.001; LES: R 2 = 0.27, p < 0.001; WSS: R 2 = 0.49, p < 0.001). The hypomethylated status of SSTR4 may involve in the development of AD and increase the risk of AD persistence in Han Chinese males.
Collapse
Affiliation(s)
- Rongrong Zhao
- The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Huihui Shi
- The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Jiajun Yin
- Brain Science Basic Laboratory, The Affiliated Wuxi Mental Health Center with Nanjing Medical University, Wuxi, China
| | - Zhen Sun
- The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yahui Xu
- The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
17
|
Yu X, Yan H, Li W. Recent advances in neuropeptide-related omics and gene editing: Spotlight on NPY and somatostatin and their roles in growth and food intake of fish. Front Endocrinol (Lausanne) 2022; 13:1023842. [PMID: 36267563 PMCID: PMC9576932 DOI: 10.3389/fendo.2022.1023842] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Feeding and growth are two closely related and important physiological processes in living organisms. Studies in mammals have provided us with a series of characterizations of neuropeptides and their receptors as well as their roles in appetite control and growth. The central nervous system, especially the hypothalamus, plays an important role in the regulation of appetite. Based on their role in the regulation of feeding, neuropeptides can be classified as orexigenic peptide and anorexigenic peptide. To date, the regulation mechanism of neuropeptide on feeding and growth has been explored mainly from mammalian models, however, as a lower and diverse vertebrate, little is known in fish regarding the knowledge of regulatory roles of neuropeptides and their receptors. In recent years, the development of omics and gene editing technology has accelerated the speed and depth of research on neuropeptides and their receptors. These powerful techniques and tools allow a more precise and comprehensive perspective to explore the functional mechanisms of neuropeptides. This paper reviews the recent advance of omics and gene editing technologies in neuropeptides and receptors and their progresses in the regulation of feeding and growth of fish. The purpose of this review is to contribute to a comparative understanding of the functional mechanisms of neuropeptides in non-mammalians, especially fish.
Collapse
|
18
|
Reassessment of SST4 Somatostatin Receptor Expression Using SST4-eGFP Knockin Mice and the Novel Rabbit Monoclonal Anti-Human SST4 Antibody 7H49L61. Int J Mol Sci 2021; 22:ijms222312981. [PMID: 34884783 PMCID: PMC8657703 DOI: 10.3390/ijms222312981] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 11/28/2021] [Accepted: 11/29/2021] [Indexed: 02/07/2023] Open
Abstract
Among the five somatostatin receptors (SST1–SST5), SST4 is the least characterized, which is in part due to the lack of specific monoclonal antibodies. We generated a knockin mouse model that expresses a carboxyl-terminal SST4-eGFP fusion protein. In addition, we extensively characterized the novel rabbit monoclonal anti-human SST4 antibody 7H49L61 using transfected cells and receptor-expressing tissues. 7H49L61 was then subjected to immunohistochemical staining of a series of formalin-fixed, paraffin-embedded normal and neoplastic human tissues. Characterization of SST4-eGFP mice revealed prominent SST4 expression in cortical pyramidal cells and trigeminal ganglion cells. In the human cortex, 7H49L61 disclosed a virtually identical staining pattern. Specificity of 7H49L61 was demonstrated by detection of a broad band migrating at 50–60 kDa in immunoblots. Tissue immunostaining was abolished by preadsorption of 7H49L61 with its immunizing peptide. In the subsequent immunohistochemical study, 7H49L61 yielded a predominant plasma membrane staining in adrenal cortex, exocrine pancreas, and placenta. SST4 was also found in glioblastomas, parathyroid adenomas, gastric and pancreatic adenocarcinomas, pheochromocytomas, and lymphomas. Altogether, we provide the first unequivocal localization of SST4 in normal and neoplastic human tissues. The monoclonal antibody 7H49L61 may also prove of great value for identifying SST4-expressing tumors during routine histopathological examinations.
Collapse
|
19
|
Gaskins DL, Burke AR, Sajdyk TJ, Truitt WA, Dietrich AD, Shekhar A. Role of Basolateral Amygdalar Somatostatin 2 Receptors in a Rat Model of Chronic Anxiety. Neuroscience 2021; 477:40-49. [PMID: 34487822 PMCID: PMC9744088 DOI: 10.1016/j.neuroscience.2021.08.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/15/2022]
Abstract
Repeated exposure to stress has been implicated in inducing chronic anxiety states. Stress related increases in anxiety responses are likely mediated by activation of corticotropin-releasing factor receptors (CRFR) in the amygdala, particularly the basolateral amygdala (BLA). Within the BLA, acute injections of the CRFR agonist urocortin 1 (Ucn1) leads to acute anxiety, whereas repeated daily injections of subthreshold-doses of Ucn1 produces a long-lasting, persistent anxiety-like phenotype, a phenomenon referred to as Ucn1-priming. Relative gene expressions from the BLA of vehicle and Ucn1-primed rats were analyzed with quantitative RT-PCR using a predesigned panel of 82 neuroscience-related genes. Compared to vehicle-primed rats, only expression of the somatostatin receptor 2 gene (Sstr2) was significantly reduced in the BLA of Ucn1-primed rats. The contribution of Sstr2 on an anxiety phenotype was tested by injecting a Sstr2 antagonist into the BLA in un-primed rats. The Sstr2 antagonist increased anxiety-like behavior. Notably, pretreatment with Sstr2 agonist injected into the BLA blocked anxiety-inducing effects of acute Ucn1 BLA-injections and delayed anxiety expression during Ucn1-priming. However, concomitant Sstr2 agonist pretreatment during Ucn-1 priming did not prevent either the development of a chronic anxiety state or a reduction of BLA Sstr2 expression induced by priming. The data demonstrate that the persistent anxiety-like phenotype observed with Ucn1-priming in the BLA is associated with a selective reduction of Sstr2 gene expression. Although Sstr2 activation in the BLA blocks acute anxiogenic effects of stress and down-regulation of BLA Sstr2, it does not suppress the long-term consequences of prolonged exposure to stress-related challenges.
Collapse
Affiliation(s)
- Denise L Gaskins
- Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 W. 15th Street, Indianapolis, IN 46202, USA; Graduate Program in Medical Neuroscience, Indiana University School of Medicine, 320 W. 15th Street, Indianapolis, IN 46202, USA
| | - Andrew R Burke
- Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 W. 15th Street, Indianapolis, IN 46202, USA; Department of Anatomy Cellular Biology & Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202, USA
| | - Tammy J Sajdyk
- Department of Psychiatry, Indiana University School of Medicine, 355 W. 16th Street, Indianapolis, IN 46202, USA; Indiana Clinical and Translational Sciences Institute, Indiana University School of Medicine, 410 W 10th St Suite 1000, Indianapolis, IN 46202, USA; Department of Pediatrics, Division of Hematology/Oncology, Indiana University School of Medicine, 705 Riley Hospital Drive, Indianapolis, IN 46202, USA
| | - William A Truitt
- Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 W. 15th Street, Indianapolis, IN 46202, USA; Department of Anatomy Cellular Biology & Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202, USA.
| | - Amy D Dietrich
- Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 W. 15th Street, Indianapolis, IN 46202, USA; Department of Anatomy Cellular Biology & Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202, USA
| | - Anantha Shekhar
- Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 W. 15th Street, Indianapolis, IN 46202, USA; Department of Psychiatry, Indiana University School of Medicine, 355 W. 16th Street, Indianapolis, IN 46202, USA; Indiana Clinical and Translational Sciences Institute, Indiana University School of Medicine, 410 W 10th St Suite 1000, Indianapolis, IN 46202, USA; Department of Psychiatry, University of Pittsburgh School of Medicine, 3550 Terrace Street, Suite 401, Pittsburgh, PA 15261, USA
| |
Collapse
|
20
|
Somatostatin receptor 4 agonism normalizes stress-related excessive amygdala glutamate release and Pavlovian aversion learning and memory in rodents. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2021; 2:470-479. [PMID: 36324659 PMCID: PMC9616361 DOI: 10.1016/j.bpsgos.2021.11.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/30/2021] [Accepted: 11/03/2021] [Indexed: 12/15/2022] Open
Abstract
Background Excessive processing of aversive life events is a major pathology in stress-related anxiety and depressive disorders. Current pharmacological treatments have rather nonspecific mechanisms of action. Somatostatin is synthesized and released as an inhibitory co-neurotransmitter by specific GABA (gamma-aminobutyric acid) interneurons, and one of its receptors, SSTR4 (somatostatin receptor 4), is localized in brain regions involved in adaptive aversion processing and implicated in negative valence neuropathology, including the amygdala. Methods Rat and mouse experiments were conducted to investigate effects of specific SSTR4 agonism on neurobehavioral aversion processing, including any normalization of stress-related hyperresponsiveness. A mouse experiment to investigate stress and SSTR4 agonism effects on reward processing was also conducted. Results In male rats (n = 5–10/group) fitted with glutamate biosensors in basolateral amygdala, SSTR4 agonism attenuated glutamate release to restraint stress in control rats and particularly in rats previously exposed to chronic corticosterone. In male mice (n = 10–18/group), SSTR4 agonism dose-dependently attenuated Pavlovian tone/footshock learning and memory measured as freezing behavior, in both control mice and mice exposed to chronic social stress, which induces excessive Pavlovian aversion learning and memory. Specificity of SSTR4 agonism effects to aversion learning/memory was demonstrated by absence of effects on discriminative reward (sucrose) learning/memory in both control mice and mice exposed to chronic social stress; SSTR4 agonism did increase reward-to-effort valuation in a dose-dependent manner and in both control mice and mice exposed to chronic social stress, which attenuates reward motivation. Conclusions These neuropsychopharmacological findings add substantially to the preclinical proof-of-concept evidence for SSTR4 agonism as a treatment in anxiety and depressive disorders.
Collapse
|
21
|
Peng L, Liu X, Yang Y, Guo Q, Zhong T. Histone Deacetylase 2-Mediated Epigenetic Regulation is Involved in the Early Isoflurane Exposure-Related Increase in Susceptibility to Anxiety-Like Behaviour Evoked by Chronic Variable Stress in Mice. Neurochem Res 2021; 46:2333-2347. [PMID: 34101131 DOI: 10.1007/s11064-021-03368-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/14/2021] [Accepted: 05/28/2021] [Indexed: 10/21/2022]
Abstract
Increasing studies report that prolonged or multiple anaesthetic exposures early in life are associated with detrimental effects on brain function. Although studies have evaluated the detrimental effects on neurocognitive function, few have focused on long-term neuropsychiatric effects. In the present study, C57BL/6 mice received either three neonatal isoflurane exposures or control exposure. Starting on postnatal day 45, the mice were either exposed or not to a chronic variable stress (CVS) paradigm, and CVS-related neuropsychiatric performance was evaluated using a series of behavioural tests. The expression levels of histone 3 lysine 9 acetylation (acetyl-H3K9), brain-derived neurotrophic factor (BDNF), cAMP response element-binding protein-binding protein, and histone deacetylases 1-4 in the amygdala were measured by immunoblotting or immunohistochemistry analysis. In mice with neonatal isoflurane exposure, the effects of sodium butyrate (NaB), a commonly used HDAC inhibitor, were examined on CVS-related behavioural and molecular alterations. The results showed that repeated neonatal isoflurane exposure did not affect innate depression-like and anxiety-like behaviours under non-stress conditions but facilitated the CVS-induced anxiety-like behavioural phenotype. Increased HDAC2 expression in the amygdala was associated with an increase in the CVS-induced repression of acetyl-H3K9 and BDNF expression and an enhanced CVS-evoked anxiety-like behavioural phenotype in mice neonatal isoflurane exposure. NaB significantly decreased the CVS-induced anxiety level by elevating acetyl-H3K9 and BDNF expression. These results suggested that early anaesthesia exposure facilitated chronic stress-induced neuropsychiatric outcomes, and the HDAC2-related epigenetic dysregulation of BDNF gene expression is involved in the underlying mechanism.
Collapse
Affiliation(s)
- Luofang Peng
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital of Central South University, Xiangya Road, 87#, Changsha City, 410008, Hunan Province, People's Republic of China
- Department of Anaesthesiology and Operating Theatre Services, Xiangya Hospital of Central South University, Xiangya Road, 87#, Changsha City, 410008, Hunan Province, People's Republic of China
| | - Xian Liu
- Department of Anaesthesiology and Operating Theatre Services, Xiangya Hospital of Central South University, Xiangya Road, 87#, Changsha City, 410008, Hunan Province, People's Republic of China
- Teaching and Research Section of Anaesthesia and Critical Care Medicine, Xiangya Hospital of Central South University, Changsha City, Hunan Province, People's Republic of China
| | - Yong Yang
- Department of Anaesthesiology and Operating Theatre Services, Xiangya Hospital of Central South University, Xiangya Road, 87#, Changsha City, 410008, Hunan Province, People's Republic of China
- Teaching and Research Section of Anaesthesia and Critical Care Medicine, Xiangya Hospital of Central South University, Changsha City, Hunan Province, People's Republic of China
| | - Qulian Guo
- Department of Anaesthesiology and Operating Theatre Services, Xiangya Hospital of Central South University, Xiangya Road, 87#, Changsha City, 410008, Hunan Province, People's Republic of China
- Teaching and Research Section of Anaesthesia and Critical Care Medicine, Xiangya Hospital of Central South University, Changsha City, Hunan Province, People's Republic of China
| | - Tao Zhong
- Department of Anaesthesiology and Operating Theatre Services, Xiangya Hospital of Central South University, Xiangya Road, 87#, Changsha City, 410008, Hunan Province, People's Republic of China.
- Teaching and Research Section of Anaesthesia and Critical Care Medicine, Xiangya Hospital of Central South University, Changsha City, Hunan Province, People's Republic of China.
| |
Collapse
|
22
|
Neumann WL, Sandoval KE, Mobayen S, Minaeian M, Kukielski SG, Srabony KN, Frare R, Slater O, Farr SA, Niehoff ML, Hospital A, Kontoyianni M, Crider AM, Witt KA. Synthesis and structure-activity relationships of 3,4,5-trisubstituted-1,2,4-triazoles: high affinity and selective somatostatin receptor-4 agonists for Alzheimer's disease treatment. RSC Med Chem 2021; 12:1352-1365. [PMID: 34458738 DOI: 10.1039/d1md00044f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/14/2021] [Indexed: 12/23/2022] Open
Abstract
Somatostatin receptor-4 (SST4) is highly expressed in brain regions affiliated with learning and memory. SST4 agonist treatment may act to mitigate Alzheimer's disease (AD) pathology. An integrated approach to SST4 agonist lead optimization is presented herein. High affinity and selective agonists with biological efficacy were identified through iterative cycles of a structure-based design strategy encompassing computational methods, chemistry, and preclinical pharmacology. 1,2,4-Triazole derivatives of our previously reported hit (4) showed enhanced SST4 binding affinity, activity, and selectivity. Thirty-five compounds showed low nanomolar range SST4 binding affinity, 12 having a K i < 1 nM. These compounds showed >500-fold affinity for SST4 as compared to SST2A. SST4 activities were consistent with the respective SST4 binding affinities (EC50 < 10 nM for 34 compounds). Compound 208 (SST4 K i = 0.7 nM; EC50 = 2.5 nM; >600-fold selectivity over SST2A) display a favorable physiochemical profile, and was advanced to learning and memory behavior evaluations in the senescence accelerated mouse-prone 8 model of AD-related cognitive decline. Chronic administration enhanced learning with i.p. dosing (1 mg kg-1) compared to vehicle. Chronic administration enhanced memory with both i.p. (0.01, 0.1, 1 mg kg-1) and oral (0.01, 10 mg kg-1) dosing compared to vehicle. This study identified a novel series of SST4 agonists with high affinity, selectivity, and biological activity that may be useful in the treatment of AD.
Collapse
Affiliation(s)
- William L Neumann
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - Karin E Sandoval
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - Shirin Mobayen
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - Mahsa Minaeian
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - Stephen G Kukielski
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - Khush N Srabony
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - Rafael Frare
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - Olivia Slater
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - Susan A Farr
- Research and Development Service, VA Medical Center, Division of Geriatric Medicine, Saint Louis University School of Medicine 1402 South Grand Boulevard, M238 St Louis MO 63104 USA
| | - Michael L Niehoff
- Research and Development Service, VA Medical Center, Division of Geriatric Medicine, Saint Louis University School of Medicine 1402 South Grand Boulevard, M238 St Louis MO 63104 USA
| | - Audrey Hospital
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - Maria Kontoyianni
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - A Michael Crider
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - Ken A Witt
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| |
Collapse
|
23
|
Weng G, Zhou B, Liu T, Huang Z, Huang S. Tetramethylpyrazine Improves Cognitive Function of Alzheimer's Disease Mice by Regulating SSTR4 Ubiquitination. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:2385-2399. [PMID: 34103899 PMCID: PMC8179737 DOI: 10.2147/dddt.s290030] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 04/12/2021] [Indexed: 01/08/2023]
Abstract
Purpose Many researches have investigated the functions of tetramethylpyrazine (TMP) in Alzheimer's disease (AD). This study aimed to discuss the underlying mechanism of TMP in AD mice. Methods TMP (200 mg/kg) was administered to 6-month-old APP/PS1 transgenic mice, and behavioral changes and hippocampal nerve injury in AD mice were detected. Apoptosis and autophagy-related protein levels were detected. Changes in gene expression before and after TMP treatment were compared using transcriptome sequencing. The effects of Cullin 4B (CUL4B) overexpression and somatostatin receptor 4 (SSTR4) silencing on AD symptoms and SSTR4 ubiquitination in APP/PS1 mice were observed. SH-SY5Y and PC12 cells were treated with 25 μmol/L Aβ25-35 and TMP to observe cell viability, apoptosis, and autophagy. Cell viability and apoptosis were measured again after treatment with proteasome inhibitor MG132 or lysosomal inhibitor 3-mA. Results TMP treatment improved the behavioral cognition of APP/PS1 mice and improved the neuronal apoptosis and damage in brain tissue. CUL4B was significantly upregulated in APP/PS1 mouse brain tissue, and SSRT4 protein was downregulated, and the levels of CUL4B and SSRT4 were negatively correlated. TMP treatment downregulated CUL4B, inhibited SSRT4 ubiquitination and upregulated SSRT4 protein level in APP/PS1 mouse brain tissue, while CUL4B overexpression or SSRT4 silencing reversed the effect of TMP. TMP and MG132 improved the decreased activity, increased apoptosis and increased SSRT4 protein in SH-SY5Y and PC12 cells treated with Aβ25-35, but not 3-mA. CUL4B overexpression promoted the ubiquitination of SSTR4 in cells, which partially reversed the effect of TMP. Conclusion TMP could improve the cognitive ability of AD mice by inhibiting CUL4B expression and the ubiquitination degradation of SSTR, and alleviating neuronal apoptosis and injury. This study may offer a new therapeutic option for AD treatment.
Collapse
Affiliation(s)
- Guohu Weng
- Department of Encephalopathy, Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou, 570203, Hainan, People's Republic of China
| | - Bo Zhou
- Department of Cardiology, Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou, 570203, Hainan, People's Republic of China
| | - Tao Liu
- Department of Neurology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, People's Republic of China
| | - Zhengxin Huang
- Department of Cardiology, Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou, 570203, Hainan, People's Republic of China
| | - Shixiong Huang
- Department of Neurology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, People's Republic of China
| |
Collapse
|
24
|
Peng L, Zhu M, Yang Y, Lu F, Liu X, Guo Q, Zhong T. Repeated Neonatal Isoflurane Exposure is Associated with Higher Susceptibility to Chronic Variable Stress-induced Behavioural and Neuro-inflammatory Alterations. Neuroscience 2021; 465:166-176. [PMID: 33951503 DOI: 10.1016/j.neuroscience.2021.04.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/11/2021] [Accepted: 04/21/2021] [Indexed: 01/22/2023]
Abstract
Numerous studies have reported that prolonged or multiple exposures to anaesthetics in early life lead to detrimental effects on brain function, most having focused on neurocognitive function, and relatively few on long term neuropsychiatric performance. The present study investigated the impact of repeated neonatal isoflurane exposure on chronic variable stress (CVS)-induced psychiatric and behavioural outcomes together with CVS-related neuronal activity and neuro-inflammatory reactivity in relevant brain circuits. In the present study, C57BL/6J mice received either three exposures to isoflurane at postnatal days 7, 8, and 9 or a control exposure. From postnatal day 45, mice were exposed to a mild, 3-week, CVS paradigm or none and the CVS-related neuropsychiatric performance was evaluated using a series of behavioural tests. The neuronal activity in relevant brain regions was measured by ΔFosB immunopositivity and CVS-related neuroinflammation was assessed by analysing levels of pro-inflammatory cytokines IL-1α, IL-1β, IL-6, and TNF-α. In mice experiencing serial neonatal isoflurane exposure, we detected a significant enhancement in anxiety levels following CVS procedures, together with enhanced neuronal activity, and exacerbated neuroinflammation in the basolateral amygdaloid nuclei (BLA) and hippocampal dentate gyrus (DG) regions. No such change was found in control mice. These results indicate an association between early multiple isoflurane exposures in infant mice and susceptibility to a CVS-evoked anxious phenotype accompanied by enhanced neuronal activity in BLA and DG regions and high inflammatory reactivity in response to CVS.
Collapse
Affiliation(s)
- Luofang Peng
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital of Central South University, Changsha City, Hunan Province, PR China; Department of Anaesthesiology and Operating Theatre Services, Xiangya Hospital of Central South University, Changsha City, Hunan Province, PR China
| | - Maoen Zhu
- Department of Anaesthesiology and Operating Theatre Services, Xiangya Hospital of Central South University, Changsha City, Hunan Province, PR China; Teaching and Research Section of Anesthesia and Critical Care Medicine, Xiangya Hospital of Central South University, Changsha City, Hunan Province, PR China
| | - Yong Yang
- Department of Anaesthesiology and Operating Theatre Services, Xiangya Hospital of Central South University, Changsha City, Hunan Province, PR China; Teaching and Research Section of Anesthesia and Critical Care Medicine, Xiangya Hospital of Central South University, Changsha City, Hunan Province, PR China
| | - Feng Lu
- Department of Anaesthesiology and Operating Theatre Services, Xiangya Hospital of Central South University, Changsha City, Hunan Province, PR China; Teaching and Research Section of Anesthesia and Critical Care Medicine, Xiangya Hospital of Central South University, Changsha City, Hunan Province, PR China
| | - Xian Liu
- Department of Anaesthesiology and Operating Theatre Services, Xiangya Hospital of Central South University, Changsha City, Hunan Province, PR China; Teaching and Research Section of Anesthesia and Critical Care Medicine, Xiangya Hospital of Central South University, Changsha City, Hunan Province, PR China
| | - Qulian Guo
- Department of Anaesthesiology and Operating Theatre Services, Xiangya Hospital of Central South University, Changsha City, Hunan Province, PR China; Teaching and Research Section of Anesthesia and Critical Care Medicine, Xiangya Hospital of Central South University, Changsha City, Hunan Province, PR China
| | - Tao Zhong
- Department of Anaesthesiology and Operating Theatre Services, Xiangya Hospital of Central South University, Changsha City, Hunan Province, PR China; Teaching and Research Section of Anesthesia and Critical Care Medicine, Xiangya Hospital of Central South University, Changsha City, Hunan Province, PR China.
| |
Collapse
|
25
|
Human Somatostatin SST 4 Receptor Transgenic Mice: Construction and Brain Expression Pattern Characterization. Int J Mol Sci 2021; 22:ijms22073758. [PMID: 33916620 PMCID: PMC8038480 DOI: 10.3390/ijms22073758] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/13/2022] Open
Abstract
Somatostatin receptor subtype 4 (SST4) has been shown to mediate analgesic, antidepressant and anti-inflammatory functions without endocrine actions; therefore, it is proposed to be a novel target for drug development. To overcome the species differences of SST4 receptor expression and function between humans and mice, we generated an SST4 humanized mouse line to serve as a translational animal model for preclinical research. A transposon vector containing the hSSTR4 and reporter gene construct driven by the hSSTR4 regulatory elements were created. The vector was randomly inserted in Sstr4-deficient mice. hSSTR4 expression was detected by bioluminescent in vivo imaging of the luciferase reporter predominantly in the brain. RT-qPCR confirmed the expression of the human gene in the brain and various peripheral tissues consistent with the in vivo imaging. RNAscope in situ hybridization revealed the presence of hSSTR4 transcripts in glutamatergic excitatory neurons in the CA1 and CA2 regions of the hippocampus; in the GABAergic interneurons in the granular layer of the olfactory bulb and in both types of neurons in the primary somatosensory cortex, piriform cortex, prelimbic cortex and amygdala. This novel SST4 humanized mouse line might enable us to investigate the differences of human and mouse SST4 receptor expression and function and assess the effects of SST4 receptor agonist drug candidates.
Collapse
|
26
|
Zhou G, Wang T, Zha XM. RNA-Seq analysis of knocking out the neuroprotective proton-sensitive GPR68 on basal and acute ischemia-induced transcriptome changes and signaling in mouse brain. FASEB J 2021; 35:e21461. [PMID: 33724568 PMCID: PMC7970445 DOI: 10.1096/fj.202002511r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/14/2021] [Accepted: 02/04/2021] [Indexed: 12/17/2022]
Abstract
Brain acid signaling plays important roles in both physiological and disease conditions. One key neuronal metabotropic proton receptor in the brain is GPR68, which contributes to hippocampal long-term potentiation (LTP) and mediates neuroprotection in acidotic and ischemic conditions. Here, to gain greater understanding of GPR68 function in the brain, we performed mRNA-Seq analysis in mice. First, we studied sham-operated animals to determine baseline expression. Compared to wild type (WT), GPR68-/- (KO) brain downregulated genes that are enriched in Gene Ontology (GO) terms of misfolding protein binding, response to organic cyclic compounds, and endoplasmic reticulum chaperone complex. Next, we examined the expression profile following transient middle cerebral artery occlusion (tMCAO). tMCAO-upregulated genes cluster to cytokine/chemokine-related functions and immune responses, while tMCAO-downregulated genes cluster to channel activities and synaptic signaling. For proton-sensitive receptors, tMCAO downregulated ASIC1a and upregulated GPR4 and GPR65, but had no effect on ASIC2, PAC, or GPR68. GPR68 deletion did not alter the expression of these proton receptors, either at baseline or after ischemia. Lastly, we performed GeneVenn analysis of differential genes at baseline and post-tMCAO. Ischemia upregulated the expression of three hemoglobin genes, along with H2-Aa, Ppbp, Siglece, and Tagln, in WT but not in KO. Immunostaining showed that tMCAO-induced hemoglobin localized to neurons. Western blot analysis further showed that hemoglobin induction is GPR68-dependent. Together, these data suggest that GPR68 deletion at baseline disrupts chaperone functions and cellular signaling responses and imply a contribution of hemoglobin-mediated antioxidant mechanism to GPR68-dependent neuroprotection in ischemia.
Collapse
Affiliation(s)
- Guokun Zhou
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL, USA
| | - Tao Wang
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL, USA
| | - Xiang-Ming Zha
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL, USA
| |
Collapse
|
27
|
Dimethyl Trisulfide Diminishes Traumatic Neuropathic Pain Acting on TRPA1 Receptors in Mice. Int J Mol Sci 2021; 22:ijms22073363. [PMID: 33806000 PMCID: PMC8036544 DOI: 10.3390/ijms22073363] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/20/2022] Open
Abstract
Pharmacotherapy of neuropathic pain is still challenging. Our earlier work indicated an analgesic effect of dimethyl trisulfide (DMTS), which was mediated by somatostatin released from nociceptor nerve endings acting on SST4 receptors. Somatostatin release occurred due to TRPA1 ion channel activation. In the present study, we investigated the effect of DMTS in neuropathic pain evoked by partial ligation of the sciatic nerve in mice. Expression of the mRNA of Trpa1 in murine dorsal-root-ganglion neurons was detected by RNAscope. Involvement of TRPA1 ion channels and SST4 receptors was tested with gene-deleted animals. Macrophage activity at the site of the nerve lesion was determined by lucigenin bioluminescence. Density and activation of microglia in the spinal cord dorsal horn was verified by immunohistochemistry and image analysis. Trpa1 mRNA is expressed in peptidergic and non-peptidergic neurons in the dorsal root ganglion. DMTS ameliorated neuropathic pain in Trpa1 and Sstr4 WT mice, but not in KO ones. DMTS had no effect on macrophage activity around the damaged nerve. Microglial density in the dorsal horn was reduced by DMTS independently from TRPA1. No effect on microglial activation was detected. DMTS might offer a novel therapeutic opportunity in the complementary treatment of neuropathic pain.
Collapse
|
28
|
Kántás B, Szőke É, Börzsei R, Bánhegyi P, Asghar J, Hudhud L, Steib A, Hunyady Á, Horváth Á, Kecskés A, Borbély É, Hetényi C, Pethő G, Pintér E, Helyes Z. In Silico, In Vitro and In Vivo Pharmacodynamic Characterization of Novel Analgesic Drug Candidate Somatostatin SST 4 Receptor Agonists. Front Pharmacol 2021; 11:601887. [PMID: 33815096 PMCID: PMC8015869 DOI: 10.3389/fphar.2020.601887] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/30/2020] [Indexed: 11/26/2022] Open
Abstract
Background: Somatostatin released from the capsaicin-sensitive sensory nerves mediates analgesic and anti-inflammatory effects via its receptor subtype 4 (SST4) without influencing endocrine functions. Therefore, SST4 is considered to be a novel target for drug development in pain, especially chronic neuropathy which is a great unmet medical need. Purpose and Experimental Approach: Here, we examined the in silico binding, SST4-linked G protein activation and β-arrestin activation on stable SST4 expressing cells and the effects of our novel pyrrolo-pyrimidine molecules (20, 100, 500, 1,000, 2,000 µg·kg−1) on partial sciatic nerve ligation-induced traumatic mononeuropathic pain model in mice. Key Results: The novel compounds bind to the high affinity binding site of SST4 the receptor and activate the G protein. However, unlike the reference SST4 agonists NNC 26-9100 and J-2156, they do not induce β-arrestin activation responsible for receptor desensitization and internalization upon chronic use. They exert 65–80% maximal anti-hyperalgesic effects in the neuropathy model 1 h after a single oral administration of 100–500 µg·kg−1 doses. Conclusion and Implications: The novel orally active compounds show potent and effective SST4 receptor agonism in vitro and in vivo. All four novel ligands proved to be full agonists based on G protein activation, but failed to recruit β-arrestin. Based on their potent antinociceptive effect in the neuropathic pain model following a single oral administration, they are promising candidates for drug development.
Collapse
Affiliation(s)
- Boglárka Kántás
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center and Center for Neuroscience, University of Pécs, Pécs, Hungary
| | - Éva Szőke
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center and Center for Neuroscience, University of Pécs, Pécs, Hungary.,PharmInVivo Ltd., Pécs, Hungary
| | - Rita Börzsei
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | | | - Junaid Asghar
- Gomal Centre of Pharmaceutical Sciences, Gomal University, Khyber Pakhtoonkhwa, Pakistan
| | - Lina Hudhud
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center and Center for Neuroscience, University of Pécs, Pécs, Hungary
| | - Anita Steib
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center and Center for Neuroscience, University of Pécs, Pécs, Hungary
| | - Ágnes Hunyady
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center and Center for Neuroscience, University of Pécs, Pécs, Hungary
| | - Ádám Horváth
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center and Center for Neuroscience, University of Pécs, Pécs, Hungary
| | - Angéla Kecskés
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center and Center for Neuroscience, University of Pécs, Pécs, Hungary
| | - Éva Borbély
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center and Center for Neuroscience, University of Pécs, Pécs, Hungary
| | - Csaba Hetényi
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center and Center for Neuroscience, University of Pécs, Pécs, Hungary
| | - Gábor Pethő
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary.,Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | - Erika Pintér
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center and Center for Neuroscience, University of Pécs, Pécs, Hungary.,PharmInVivo Ltd., Pécs, Hungary.,Algonist Biotechnolgies GmbH, Vienna, Austria
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center and Center for Neuroscience, University of Pécs, Pécs, Hungary.,PharmInVivo Ltd., Pécs, Hungary.,Algonist Biotechnolgies GmbH, Vienna, Austria
| |
Collapse
|
29
|
Albrecht A, Redavide E, Regev-Tsur S, Stork O, Richter-Levin G. Hippocampal GABAergic interneurons and their co-localized neuropeptides in stress vulnerability and resilience. Neurosci Biobehav Rev 2020; 122:229-244. [PMID: 33188820 DOI: 10.1016/j.neubiorev.2020.11.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/05/2020] [Accepted: 11/01/2020] [Indexed: 12/13/2022]
Abstract
Studies in humans and rodents suggest a critical role for the hippocampal formation in cognition and emotion, but also in the adaptation to stressful events. Successful stress adaptation promotes resilience, while its failure may lead to stress-induced psychopathologies such as depression and anxiety disorders. Hippocampal architecture and physiology is shaped by its strong control of activity via diverse classes of inhibitory interneurons that express typical calcium binding proteins and neuropeptides. Celltype-specific opto- and chemogenetic intervention strategies that take advantage of these biochemical markers have bolstered our understanding of the distinct role of different interneurons in anxiety, fear and stress adaptation. Moreover, some of the signature proteins of GABAergic interneurons have a potent impact on emotion and cognition on their own, making them attractive targets for interventions. In particular, neuropeptide Y is a promising endogenous agent for mediating resilience against severe stress. In this review, we evaluate the role of the major types of interneurons across hippocampal subregions in the adaptation to chronic and acute stress and to emotional memory formation.
Collapse
Affiliation(s)
- Anne Albrecht
- Institute of Anatomy, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany; Center for Behavioral Brain Science, Universitätsplatz 2, 39106 Magdeburg, Germany.
| | - Elisa Redavide
- Institute of Anatomy, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany; Center for Behavioral Brain Science, Universitätsplatz 2, 39106 Magdeburg, Germany; Department of Genetics & Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany; Institute of Pharmacology and Toxicology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany.
| | - Stav Regev-Tsur
- Sagol Department of Neurobiology, University of Haifa, 199 Aba-Hushi Avenue, 3498838 Haifa, Israel; The Integrated Brain and Behavior Research Center (IBBR), University of Haifa, 199 Aba-Hushi Avenue, 3498838 Haifa, Israel.
| | - Oliver Stork
- Center for Behavioral Brain Science, Universitätsplatz 2, 39106 Magdeburg, Germany; Department of Genetics & Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany.
| | - Gal Richter-Levin
- Sagol Department of Neurobiology, University of Haifa, 199 Aba-Hushi Avenue, 3498838 Haifa, Israel; The Integrated Brain and Behavior Research Center (IBBR), University of Haifa, 199 Aba-Hushi Avenue, 3498838 Haifa, Israel; Psychology Department, University of Haifa199 Aba-Hushi Avenue, 3498838 Haifa, Israel.
| |
Collapse
|
30
|
Kecskés A, Pohóczky K, Kecskés M, Varga ZV, Kormos V, Szőke É, Henn-Mike N, Fehér M, Kun J, Gyenesei A, Renner É, Palkovits M, Ferdinandy P, Ábrahám IM, Gaszner B, Helyes Z. Characterization of Neurons Expressing the Novel Analgesic Drug Target Somatostatin Receptor 4 in Mouse and Human Brains. Int J Mol Sci 2020; 21:E7788. [PMID: 33096776 PMCID: PMC7589422 DOI: 10.3390/ijms21207788] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/08/2020] [Accepted: 10/16/2020] [Indexed: 12/13/2022] Open
Abstract
Somatostatin is an important mood and pain-regulating neuropeptide, which exerts analgesic, anti-inflammatory, and antidepressant effects via its Gi protein-coupled receptor subtype 4 (SST4) without endocrine actions. SST4 is suggested to be a unique novel drug target for chronic neuropathic pain, and depression, as a common comorbidity. However, its neuronal expression and cellular mechanism are poorly understood. Therefore, our goals were (i) to elucidate the expression pattern of Sstr4/SSTR4 mRNA, (ii) to characterize neurochemically, and (iii) electrophysiologically the Sstr4/SSTR4-expressing neuronal populations in the mouse and human brains. Here, we describe SST4 expression pattern in the nuclei of the mouse nociceptive and anti-nociceptive pathways as well as in human brain regions, and provide neurochemical and electrophysiological characterization of the SST4-expressing neurons. Intense or moderate SST4 expression was demonstrated predominantly in glutamatergic neurons in the major components of the pain matrix mostly also involved in mood regulation. The SST4 agonist J-2156 significantly decreased the firing rate of layer V pyramidal neurons by augmenting the depolarization-activated, non-inactivating K+ current (M-current) leading to remarkable inhibition. These are the first translational results explaining the mechanisms of action of SST4 agonists as novel analgesic and antidepressant candidates.
Collapse
Affiliation(s)
- Angéla Kecskés
- Department of Pharmacology and Pharmacotherapy, Medical School & Szentágothai Research Centre, Molecular Pharmacology Research Group, University of Pécs, H-7624 Pécs, Hungary; (A.K.); (K.P.); (V.K.); (É.S.); (J.K.)
- Centre for Neuroscience, University of Pécs, H-7624 Pécs, Hungary; (M.K.); (N.H.-M.); (I.M.Á.)
| | - Krisztina Pohóczky
- Department of Pharmacology and Pharmacotherapy, Medical School & Szentágothai Research Centre, Molecular Pharmacology Research Group, University of Pécs, H-7624 Pécs, Hungary; (A.K.); (K.P.); (V.K.); (É.S.); (J.K.)
- Centre for Neuroscience, University of Pécs, H-7624 Pécs, Hungary; (M.K.); (N.H.-M.); (I.M.Á.)
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, H-7624 Pécs, Hungary
| | - Miklós Kecskés
- Centre for Neuroscience, University of Pécs, H-7624 Pécs, Hungary; (M.K.); (N.H.-M.); (I.M.Á.)
- Institute of Physiology, Medical School & Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary
| | - Zoltán V. Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary; (Z.V.V.); (P.F.)
- HCEMM-SU Cardiometabolic Immunology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Viktória Kormos
- Department of Pharmacology and Pharmacotherapy, Medical School & Szentágothai Research Centre, Molecular Pharmacology Research Group, University of Pécs, H-7624 Pécs, Hungary; (A.K.); (K.P.); (V.K.); (É.S.); (J.K.)
- Centre for Neuroscience, University of Pécs, H-7624 Pécs, Hungary; (M.K.); (N.H.-M.); (I.M.Á.)
| | - Éva Szőke
- Department of Pharmacology and Pharmacotherapy, Medical School & Szentágothai Research Centre, Molecular Pharmacology Research Group, University of Pécs, H-7624 Pécs, Hungary; (A.K.); (K.P.); (V.K.); (É.S.); (J.K.)
- Centre for Neuroscience, University of Pécs, H-7624 Pécs, Hungary; (M.K.); (N.H.-M.); (I.M.Á.)
- ALGONIST Biotechnologies GmbH, A-1030 Wien, Austria
| | - Nóra Henn-Mike
- Centre for Neuroscience, University of Pécs, H-7624 Pécs, Hungary; (M.K.); (N.H.-M.); (I.M.Á.)
- Institute of Physiology, Medical School & Szentágothai Research Centre, PTE-NAP Molecular Neuroendocrinology Research Group, University of Pécs, H-7624 Pécs, Hungary
| | - Máté Fehér
- Department of Neurosurgery, Kaposi Mór Teaching Hospital, H-7400 Kaposvár, Hungary;
| | - József Kun
- Department of Pharmacology and Pharmacotherapy, Medical School & Szentágothai Research Centre, Molecular Pharmacology Research Group, University of Pécs, H-7624 Pécs, Hungary; (A.K.); (K.P.); (V.K.); (É.S.); (J.K.)
- Centre for Neuroscience, University of Pécs, H-7624 Pécs, Hungary; (M.K.); (N.H.-M.); (I.M.Á.)
- Bioinformatics Research Group, Genomics and Bioinformatics Core Facility, Szentágothai Research Centre University of Pécs, H-7624 Pécs, Hungary;
| | - Attila Gyenesei
- Bioinformatics Research Group, Genomics and Bioinformatics Core Facility, Szentágothai Research Centre University of Pécs, H-7624 Pécs, Hungary;
| | - Éva Renner
- Human Brain Tissue Bank, Semmelweis University, H-1089 Budapest, Hungary; (É.R.); (M.P.)
| | - Miklós Palkovits
- Human Brain Tissue Bank, Semmelweis University, H-1089 Budapest, Hungary; (É.R.); (M.P.)
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary; (Z.V.V.); (P.F.)
- Pharmahungary Group, H-6720 Szeged, Hungary
| | - István M. Ábrahám
- Centre for Neuroscience, University of Pécs, H-7624 Pécs, Hungary; (M.K.); (N.H.-M.); (I.M.Á.)
- Institute of Physiology, Medical School & Szentágothai Research Centre, PTE-NAP Molecular Neuroendocrinology Research Group, University of Pécs, H-7624 Pécs, Hungary
| | - Balázs Gaszner
- Centre for Neuroscience, University of Pécs, H-7624 Pécs, Hungary; (M.K.); (N.H.-M.); (I.M.Á.)
- Department of Anatomy, Medical School, Research Group for Mood Disorders, University of Pécs, H-7624 Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School & Szentágothai Research Centre, Molecular Pharmacology Research Group, University of Pécs, H-7624 Pécs, Hungary; (A.K.); (K.P.); (V.K.); (É.S.); (J.K.)
- Centre for Neuroscience, University of Pécs, H-7624 Pécs, Hungary; (M.K.); (N.H.-M.); (I.M.Á.)
- ALGONIST Biotechnologies GmbH, A-1030 Wien, Austria
- PharmInVivo Ltd., H-7629 Pécs, Hungary
| |
Collapse
|
31
|
Szőke É, Bálint M, Hetényi C, Markovics A, Elekes K, Pozsgai G, Szűts T, Kéri G, Őrfi L, Sándor Z, Szolcsányi J, Pintér E, Helyes Z. Small molecule somatostatin receptor subtype 4 (sst 4) agonists are novel anti-inflammatory and analgesic drug candidates. Neuropharmacology 2020; 178:108198. [PMID: 32739276 DOI: 10.1016/j.neuropharm.2020.108198] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 06/07/2020] [Accepted: 06/10/2020] [Indexed: 12/30/2022]
Abstract
We provided strong proof of concept evidence that somatostatin mediates potent analgesic and anti-inflammatory actions via its receptor subtype 4 (sst4) located both at the periphery and the central nervous system. Therefore, sst4 agonists are promising novel drug candidates for neuropathic pain and neurogenic inflammation, but rational drug design was not possible due to the lack of knowledge about its 3-dimensional structure. We modeled the sst4 receptor structure, described its agonist binding properties, and characterized the binding of our novel small molecule sst4 agonists (4-phenetylamino-7H-pyrrolo[2,3-d]pyrimidine derivatives) using an in silico platform. In addition to the in silico binding data, somatostatin displacement by Compound 1 was demonstrated in the competitive binding assay on sst4-expressing cells. In vivo effects were investigated in rat models of neurogenic inflammation and chronic traumatic neuropathic pain. We defined high- and low-affinity binding pockets of sst4 for our ligands, binding of the highest affinity compounds were similar to that of the reference ligand J-2156. We showed potent G-protein activation with the highest potency of 10 nM EC50 value and highest efficacy of 342%. Oral administration of 100 μg/kg of 5 compounds significantly inhibited acute neurogenic plasma protein extravasation in the paw skin by 40-60%, one candidate abolished and 3 others diminished sciatic nerve-ligation induced neuropathic hyperalgesia by 28-62%. The in silico predictions on sst4-ligands were tested in biological systems. Low oral dose of our novel agonists inhibit neurogenic inflammation and neuropathic pain, which opens promising drug developmental perspectives for these unmet medical need conditions.
Collapse
Affiliation(s)
- Éva Szőke
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary; János Szentágothai Research Center & Centre for Neuroscience, University of Pécs, Hungary.
| | - Mónika Bálint
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary; János Szentágothai Research Center & Centre for Neuroscience, University of Pécs, Hungary
| | - Csaba Hetényi
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary; János Szentágothai Research Center & Centre for Neuroscience, University of Pécs, Hungary
| | - Adrienn Markovics
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary; János Szentágothai Research Center & Centre for Neuroscience, University of Pécs, Hungary
| | - Krisztián Elekes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary; János Szentágothai Research Center & Centre for Neuroscience, University of Pécs, Hungary
| | - Gábor Pozsgai
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary; János Szentágothai Research Center & Centre for Neuroscience, University of Pécs, Hungary
| | | | - György Kéri
- Vichem Chemie Research Ltd, Budapest, Hungary
| | - László Őrfi
- Department of Pharmaceutical Chemistry, Pharmacy Faculty, Semmelweis University, Budapest, Hungary
| | - Zoltán Sándor
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary; János Szentágothai Research Center & Centre for Neuroscience, University of Pécs, Hungary
| | - János Szolcsányi
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary; János Szentágothai Research Center & Centre for Neuroscience, University of Pécs, Hungary
| | - Erika Pintér
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary; János Szentágothai Research Center & Centre for Neuroscience, University of Pécs, Hungary; PharmInVivo Ltd, Pécs, Hungary; Algonist GmbH, Vienna, Austria
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary; János Szentágothai Research Center & Centre for Neuroscience, University of Pécs, Hungary; PharmInVivo Ltd, Pécs, Hungary; Algonist GmbH, Vienna, Austria
| |
Collapse
|
32
|
Daniels S, Horman T, Lapointe T, Melanson B, Storace A, Kennedy SH, Frey BN, Rizvi SJ, Hassel S, Mueller DJ, Parikh SV, Lam RW, Blier P, Farzan F, Giacobbe P, Milev R, Placenza F, Soares CN, Turecki G, Uher R, Leri F. Reverse translation of major depressive disorder symptoms: A framework for the behavioural phenotyping of putative biomarkers. J Affect Disord 2020; 263:353-366. [PMID: 31969265 DOI: 10.1016/j.jad.2019.11.108] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/13/2019] [Accepted: 11/22/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Reverse translating putative biomarkers of depression from patients to animals is complex because Major Depressive Disorder (MDD) is a highly heterogenous condition. This review proposes an approach to reverse translation based on relating relevant bio-behavioural functions in laboratory rodents to MDD symptoms. METHODS This systematic review outlines symptom clusters assessed by psychometric tests of MDD and antidepressant treatment response including the Montgomery-Åsberg Depression Rating Scale, the Hamilton Depression Rating Scale, and the Beck Depression Inventory. Symptoms were related to relevant behavioural assays in laboratory rodents. RESULTS The resulting battery of tests includes passive coping, anxiety-like behaviours, sleep, caloric intake, cognition, psychomotor functions, hedonic reactivity and aversive learning. These assays are discussed alongside relevant clinical symptoms of MDD, providing a framework through which reverse translation of a biomarker can be interpreted. LIMITATIONS Certain aspects of MDD may not be quantified by tests in laboratory rodents, and their biological significance may not always be of clinical relevance. CONCLUSIONS Using this reverse translation approach, it is possible to clarify the functional significance of a putative biomarker in rodents and hence translate its contribution to specific clinical symptoms, or clusters of symptoms.
Collapse
Affiliation(s)
- Stephen Daniels
- Department of Psychology and Neuroscience, University of Guelph, Guelph N1G 2W1, Ontario, Canada
| | - Thomas Horman
- Department of Psychology and Neuroscience, University of Guelph, Guelph N1G 2W1, Ontario, Canada
| | - Thomas Lapointe
- Department of Psychology and Neuroscience, University of Guelph, Guelph N1G 2W1, Ontario, Canada
| | - Brett Melanson
- Department of Psychology and Neuroscience, University of Guelph, Guelph N1G 2W1, Ontario, Canada
| | - Alexandra Storace
- Department of Psychology and Neuroscience, University of Guelph, Guelph N1G 2W1, Ontario, Canada
| | - Sidney H Kennedy
- University of Toronto Health Network, Toronto, Ontario, Canada; St. Michael's Hospital, Toronto, Ontario, Canada
| | | | - Sakina J Rizvi
- University of Toronto Health Network, Toronto, Ontario, Canada; St. Michael's Hospital, Toronto, Ontario, Canada
| | | | - Daniel J Mueller
- The Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | | | - Raymond W Lam
- The University of British Columbia, Vancouver, British Columbia, Canada
| | - Pierre Blier
- The Royal Institute of Mental Health Research, Ottawa, Ontario, Canada
| | - Faranak Farzan
- Simon Fraser University, Burnaby, British Columbia, Canada
| | - Peter Giacobbe
- University of Toronto Health Network, Toronto, Ontario, Canada
| | | | - Franca Placenza
- University of Toronto Health Network, Toronto, Ontario, Canada
| | | | | | - Rudolf Uher
- Dalhousie University, Halifax, Nova Scotia, Canada
| | - Francesco Leri
- Department of Psychology and Neuroscience, University of Guelph, Guelph N1G 2W1, Ontario, Canada.
| |
Collapse
|
33
|
Robinson SL, Thiele TE. A role for the neuropeptide somatostatin in the neurobiology of behaviors associated with substances abuse and affective disorders. Neuropharmacology 2020; 167:107983. [PMID: 32027909 DOI: 10.1016/j.neuropharm.2020.107983] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/07/2020] [Accepted: 01/30/2020] [Indexed: 02/06/2023]
Abstract
In recent years, neuropeptides which display potent regulatory control of stress-related behaviors have been extensively demonstrated to play a critical role in regulating behaviors associated with substance abuse and affective disorders. Somatostatin (SST) is one neuropeptide known to significantly contribute to emotionality and stress behaviors. However, the role of SST in regulating behavior has received relatively little attention relative to other stress-involved peptides, such as neuropeptide Y or corticotrophin releasing factor. This review characterizes our current understanding of the role of SST and SST-expressing cells in general in modulating several behaviors intrinsically linked to substance abuse and affective disorders, specifically: anxiety and fear; stress and depression; feeding and drinking; and circadian rhythms. We further summarize evidence of a direct role for the SST system, and specifically somatostatin receptors 2 and 4, in substance abuse disorders. This article is part of the special issue on 'Neuropeptides'.
Collapse
Affiliation(s)
- Stacey L Robinson
- Department of Psychology & Neuroscience, University of North Carolina, Chapel Hill, NC, 27599, USA; Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Todd E Thiele
- Department of Psychology & Neuroscience, University of North Carolina, Chapel Hill, NC, 27599, USA; Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
34
|
Somatostatin receptors (SSTR1-5) on inhibitory interneurons in the barrel cortex. Brain Struct Funct 2019; 225:387-401. [PMID: 31873798 PMCID: PMC6957562 DOI: 10.1007/s00429-019-02011-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 12/11/2019] [Indexed: 12/11/2022]
Abstract
Inhibitory interneurons in the cerebral cortex contain specific proteins or peptides characteristic for a certain interneuron subtype. In mice, three biochemical markers constitute non-overlapping interneuron populations, which account for 80–90% of all inhibitory cells. These interneurons express parvalbumin (PV), somatostatin (SST), or vasoactive intestinal peptide (VIP). SST is not only a marker of a specific interneuron subtype, but also an important neuropeptide that participates in numerous biochemical and signalling pathways in the brain via somatostatin receptors (SSTR1-5). In the nervous system, SST acts as a neuromodulator and neurotransmitter affecting, among others, memory, learning, and mood. In the sensory cortex, the co-localisation of GABA and SST is found in approximately 30% of interneurons. Considering the importance of interactions between inhibitory interneurons in cortical plasticity and the possible GABA and SST co-release, it seems important to investigate the localisation of different SSTRs on cortical interneurons. Here, we examined the distribution of SSTR1-5 on barrel cortex interneurons containing PV, SST, or VIP. Immunofluorescent staining using specific antibodies was performed on brain sections from transgenic mice that expressed red fluorescence in one specific interneuron subtype (PV-Ai14, SST-Ai14, and VIP-Ai14 mice). SSTRs expression on PV, SST, and VIP interneurons varied among the cortical layers and we found two patterns of SSTRs distribution in L4 of barrel cortex. We also demonstrated that, in contrast to other interneurons, PV cells did not express SSTR2, but expressed other SSTRs. SST interneurons, which were not found to make chemical synapses among themselves, expressed all five SSTR subtypes.
Collapse
|
35
|
Szentes N, Tékus V, Mohos V, Borbély É, Helyes Z. Exploratory and locomotor activity, learning and memory functions in somatostatin receptor subtype 4 gene-deficient mice in relation to aging and sex. GeroScience 2019; 41:631-641. [PMID: 30903571 PMCID: PMC6885027 DOI: 10.1007/s11357-019-00059-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/08/2019] [Indexed: 02/06/2023] Open
Abstract
The inhibitory neuropeptide somatostatin regulates several functions in the nervous system including memory. Its concentrations decrease by age leading to functional alterations, but there are little known about the receptorial mechanism. We discovered that somatostatin receptor 4 (sst4) mediates analgesic, anti-depressant, and anti-inflammatory effects without endocrine actions, and it is a unique target for drug development. We investigated the exploratory and locomotor activities and learning and memory functions of male and female sst4gene-deficient mice compared with their wild-types (WT) at ages of 3, 12, 17 months in the Y-maze test, open field test (OFT), radial-arm maze (RAM) test and novel object recognition (NOR) test. Young sst4 gene-deficient females visited, repeated, and missed significantly less arms than the WTs in the RAM; males showed decreased exploration in the NOR. Young mice moved significantly more, spend longer time in OFT center, and visited more arms in the Y-maze than older ones. Young WT females spend significantly longer time in the OFT center, visited, missed and repeated more arms of the RAM than males. Old males found more rewards than females. Young males explored longer the novel object than young females and older males in the NOR; the recognition index was smaller in females. We conclude that aging and sex are important factors of behavioral parameters that should be focused on in such studies. Sst4 is likely to influence locomotion and exploratory behavior only in young mice, but not during normal aging, which is a beneficial feature of a good drug target focusing on the elderly.
Collapse
Affiliation(s)
- Nikolett Szentes
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, János Szentágothai Research Centre & Centre for Neuroscience, University of Pécs, Szigeti u. 12, Pécs, H-7624, Hungary
| | - Valéria Tékus
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, János Szentágothai Research Centre & Centre for Neuroscience, University of Pécs, Szigeti u. 12, Pécs, H-7624, Hungary
| | - Violetta Mohos
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | - Éva Borbély
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, János Szentágothai Research Centre & Centre for Neuroscience, University of Pécs, Szigeti u. 12, Pécs, H-7624, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, János Szentágothai Research Centre & Centre for Neuroscience, University of Pécs, Szigeti u. 12, Pécs, H-7624, Hungary. .,PharmInVivo Ltd., Pécs, Hungary.
| |
Collapse
|
36
|
Günther T, Tulipano G, Dournaud P, Bousquet C, Csaba Z, Kreienkamp HJ, Lupp A, Korbonits M, Castaño JP, Wester HJ, Culler M, Melmed S, Schulz S. International Union of Basic and Clinical Pharmacology. CV. Somatostatin Receptors: Structure, Function, Ligands, and New Nomenclature. Pharmacol Rev 2018; 70:763-835. [PMID: 30232095 PMCID: PMC6148080 DOI: 10.1124/pr.117.015388] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Somatostatin, also known as somatotropin-release inhibitory factor, is a cyclopeptide that exerts potent inhibitory actions on hormone secretion and neuronal excitability. Its physiologic functions are mediated by five G protein-coupled receptors (GPCRs) called somatostatin receptor (SST)1-5. These five receptors share common structural features and signaling mechanisms but differ in their cellular and subcellular localization and mode of regulation. SST2 and SST5 receptors have evolved as primary targets for pharmacological treatment of pituitary adenomas and neuroendocrine tumors. In addition, SST2 is a prototypical GPCR for the development of peptide-based radiopharmaceuticals for diagnostic and therapeutic interventions. This review article summarizes findings published in the last 25 years on the physiology, pharmacology, and clinical applications related to SSTs. We also discuss potential future developments and propose a new nomenclature.
Collapse
Affiliation(s)
- Thomas Günther
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Giovanni Tulipano
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Pascal Dournaud
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Corinne Bousquet
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Zsolt Csaba
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Hans-Jürgen Kreienkamp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Márta Korbonits
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Justo P Castaño
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Hans-Jürgen Wester
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Michael Culler
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Shlomo Melmed
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| |
Collapse
|
37
|
Kaufmann D, Brennan KC. The Effects of Chronic Stress on Migraine Relevant Phenotypes in Male Mice. Front Cell Neurosci 2018; 12:294. [PMID: 30283302 PMCID: PMC6156251 DOI: 10.3389/fncel.2018.00294] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 08/16/2018] [Indexed: 12/14/2022] Open
Abstract
Migraine is a disabling neurological disorder affecting 12% of the world’s population. Stress is a major reported trigger and exacerbator of migraine. We evaluated the effects of two chronic stress paradigms on migraine relevant phenotypes in male C57Bl/6 mice. Methods: Fifty six mice were used in a 14 day social defeat stress (SDS) and twenty three mice were used in a 40 day chronic variable stress (CVS) paradigm. Anxiety measures were evaluated using the open field and elevated plus maze (EPM) tests. Migraine relevant phenotypes were evaluated using the nitroglycerin (NTG) and cortical spreading depression (CSD) models. Results: Stress sensitive SDS mice and chronically stressed CVS mice showed decreased exploration in the open field and reduced time spent in the open arms of the EPM compared to controls. Stress sensitive and resilient SDS mice had increased serum corticosterone levels, and stressed mice in the CVS paradigm had decreased weight gain compared to controls, providing combined behavioral and physiological evidence of a stress response. In the CVS paradigm but not the SDS paradigm, the stressed group showed a significant decrease in baseline mechanical withdrawal threshold compared to controls. All groups showed a significant reduction in withdrawal threshold after treatment with NTG, but the reduction was not larger in SDS or CVS than in controls. Interestingly, stress resilient SDS mice showed a rapid recovery from NTG effects that was not seen in other groups. No difference in CSD frequency or velocity was seen between stress and control mice in either stress paradigms. Conclusion: We observed distinct effects of stress on generalized pain response, migraine relevant pain, and migraine relevant excitability. CVS but not SDS was associated with a reduced mechanical withdrawal threshold, consistent with a generalized pain response to chronic stress. Neither SDS nor CVS exacerbated phenotypes considered specifically relevant to migraine - withdrawal to NTG, and susceptibility to CSD. However, the significantly reduced response of stress resilient mice to the NTG stimulus may represent a specific migraine-resistant phenotype.
Collapse
Affiliation(s)
- Dan Kaufmann
- Headache Physiology Lab, Department of Neurology, University of Utah, Salt Lake City, UT, United States
| | - K C Brennan
- Headache Physiology Lab, Department of Neurology, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
38
|
Common variants on 6q16.2, 12q24.31 and 16p13.3 are associated with major depressive disorder. Neuropsychopharmacology 2018; 43:2146-2153. [PMID: 29728651 PMCID: PMC6098070 DOI: 10.1038/s41386-018-0078-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 04/10/2018] [Accepted: 04/17/2018] [Indexed: 12/12/2022]
Abstract
Accumulating evidence suggests that genetic factors have a role in major depressive disorder (MDD). However, only limited MDD risk loci have been identified so far. Here we perform a meta-analysis (a total of 90,150 MDD cases and 246,603 controls) through combing three genome-wide association studies of MDD, including 23andMe (cases were self-reported with a clinical diagnosis or treatment of depression), CONVERGE (cases were diagnosed using the Composite International Diagnostic Interview) and PGC (cases were diagnosed using direct structured diagnostic interview (by trained interviewers) or clinician-administered DSM-IV checklists). Genetic variants from two previously unreported loci (rs10457592 on 6q16.2 and rs2004910 on 12q24.31) showed significant associations with MDD (P < 5 × 10-8) in a total of 336,753 subjects. SNPs (a total of 171) with a P < 1 × 10-7 in the meta-analysis were further replicated in an independent sample (GS:SFHS, 2,659 MDD cases (diagnosed with DSM-IV) and 17,237 controls) and one additional risk locus (rs3785234 on 16p13.3, P = 1.57 × 10-8) was identified in the combined samples (a total of 92,809 cases and 263,840 controls). Risk variants on the identified risk loci were associated with gene expression in human brain tissues and mRNA expression analysis showed that FBXL4 and RSRC1 were significantly upregulated in brains of MDD cases compared with controls, suggesting that genetic variants may confer risk of MDD through regulating the expression of these two genes. Our study identified three novel risk loci (6q16.2, 12q24.31, and 16p13.3) for MDD and suggested that FBXL4 and RSRC1 may play a role in MDD. Further functional characterization of the identified risk genes may provide new insights for MDD pathogenesis.
Collapse
|
39
|
Prévôt TD, Viollet C, Epelbaum J, Dominguez G, Béracochéa D, Guillou JL. sst 2-receptor gene deletion exacerbates chronic stress-induced deficits: Consequences for emotional and cognitive ageing. Prog Neuropsychopharmacol Biol Psychiatry 2018; 86:390-400. [PMID: 29409919 DOI: 10.1016/j.pnpbp.2018.01.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/17/2018] [Accepted: 01/30/2018] [Indexed: 02/07/2023]
Abstract
This study investigated whether sst2 gene deletion interacts with age and chronic stress exposure to produce exacerbated emotional and cognitive ageing. Middle-aged (10-12 month) sst2 knockout (sst2KO) and wild-type (WT) mice underwent an unpredictable chronic mild stress (UCMS) procedure for 6 weeks or no stress for control groups. This was followed by a battery of tests to assess emotional and cognitive functions and neuroendocrine status (CORT level). A re-evaluation was performed 6 months later (i.e. with 18-month-old mice). UCMS reproduced neuroendocrine and behavioral features of stress-related disorders such as elevated circulating CORT levels, physical deteriorations, increased anxiety- and depressive-like behaviors and working memory impairments. sst2KO mice displayed behavioral alterations which were similar to stressed WT and exhibited exacerbated changes following UCMS exposure. The evaluations performed in the older mice showed significant long-term effects of UCMS exposure. Old sst2KO mice previously exposed to UCMS exhibited spatial learning and memory accuracy impairments and high levels of anxiety-like behaviors which drastically added to the effects of normal ageing. Spatial abilities and emotionality scores (mean z-scores) measured both at the UCMS outcome and 6 months later were correlated with the initially measured CORT levels in middle-age. The present findings indicate that the deletion of the sst2 receptor gene produces chronic hypercorticosteronemia and exacerbates sensitivity to stressors which over time, have consequences on ageing brain function processes.
Collapse
Affiliation(s)
- Thomas Damien Prévôt
- Université de Bordeaux, Pessac, France; Centre National de la Recherche Scientifique, UMR 5287, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Pessac, France
| | - Cécile Viollet
- Inserm, UMR 894, Center for Psychiatry & Neuroscience, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jacques Epelbaum
- Inserm, UMR 894, Center for Psychiatry & Neuroscience, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; UMR 7179 CNRS MNHN - MECADEV, 91800 Brunoy, France
| | - Gaëlle Dominguez
- Centre National de la Recherche Scientifique, UMR 5287, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Pessac, France
| | - Daniel Béracochéa
- Université de Bordeaux, Pessac, France; Centre National de la Recherche Scientifique, UMR 5287, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Pessac, France
| | - Jean-Louis Guillou
- Université de Bordeaux, Pessac, France; Centre National de la Recherche Scientifique, UMR 5287, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Pessac, France.
| |
Collapse
|