1
|
Chen X, Chen Y, Shu R, Lu S, Gu MM, Shen C, Wang Z, Cui X. Investigating the effects of global gene knockout of MrgF on motor performance and pain sensitivity in mice. Hereditas 2025; 162:31. [PMID: 40033362 PMCID: PMC11874108 DOI: 10.1186/s41065-025-00377-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 01/25/2025] [Indexed: 03/05/2025] Open
Abstract
Mas-related G protein-coupled receptors (Mrgs) are a subset of GPCRs linked to pain modulation. MrgF was identified as an orphan Mrg whose function and ligand remain unclear. In this study, in addition to its expression in the dorsal root ganglia (DRG), the primary afferent center that transmits pain, we identified dense expression of MrgF, particularly concentrated in the Purkinje cell layer of the mouse cerebellum. Given the the important role of Purkinje neurons in both motor modulation and non-motor modulation, including pain processing, we established a MrgF knockout mouse (MrgF-/-) model and performed a battery of behavioral tests to explore motor performance and assess pain-associated responses. MrgF-/- mice exhibited no disturbances in coordination and motor balance during the rotarod, pole, balance beam, and treadmill tests, and normal cerebellar histology was retained. In hot plate assays, MrgF-/- mice displayed reduced pain-related behavioral responses to thermal stimuli, although no significance differences were found in tail flick assays between MrgF-/- and wild-type (wt) mice. Moreover, in formalin tests, MrgF-/- mice also showed decreased chemical-induced nociception. This was accompanied by a downregulation in the expression levels of genes associated with nociceptive modulation, such as c-fos, Runx1, Nav1.7, Nav1.8, and Nav1.9, within the DRG of MrgF-/- mice. Taken together, our findings suggest that MrgF may play a significant role in modulating pain sensitivity, thereby advancing the understanding of the functional characteristics of the Mrgs family.
Collapse
Affiliation(s)
- Xuejiao Chen
- Institute of Neuroscience and Brain science, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China.
| | - Yan Chen
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Runzhe Shu
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shunyuan Lu
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ming-Min Gu
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chunling Shen
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhugang Wang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xiaofang Cui
- Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
2
|
Banerjee D, Sultana S, Banerjee S. Gas5 regulates early-life stress-induced anxiety and spatial memory. J Neurochem 2024; 168:2999-3018. [PMID: 38960403 DOI: 10.1111/jnc.16167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 07/05/2024]
Abstract
Early-life stress (ES) induced by maternal separation (MS) remains a proven causality of anxiety and memory deficits at later stages of life. Emerging studies have shown that MS-induced gene expression in the hippocampus is operated at the level of transcription. However, the extent of involvement of non-coding RNAs in MS-induced behavioural deficits remains unexplored. Here, we have investigated the role of synapse-enriched long non-coding RNAs (lncRNAs) in anxiety and memory upon MS. We observed that MS led to an enhancement of expression of the lncRNA growth arrest specific 5 (Gas5) in the hippocampus; accompanied by increased levels of anxiety and deficits in spatial memory. Gas5 knockdown in early life was able to reduce anxiety and partially rescue the spatial memory deficits of maternally separated adult mice. However, the reversal of MS-induced anxiety and memory deficits is not attributed to Gas5 activity during neuronal development as Gas5 RNAi did not influence spine development. Gene Ontology analysis revealed that Gas5 exerts its function by regulating RNA metabolism and translation. Our study highlights the importance of MS-regulated lncRNA in anxiety and spatial memory.
Collapse
Affiliation(s)
| | - Sania Sultana
- National Brain Research Centre, Gurugram, Haryana, India
| | | |
Collapse
|
3
|
Birgül Iyison N, Abboud C, Abboud D, Abdulrahman AO, Bondar AN, Dam J, Georgoussi Z, Giraldo J, Horvat A, Karoussiotis C, Paz-Castro A, Scarpa M, Schihada H, Scholz N, Güvenc Tuna B, Vardjan N. ERNEST COST action overview on the (patho)physiology of GPCRs and orphan GPCRs in the nervous system. Br J Pharmacol 2024. [PMID: 38825750 DOI: 10.1111/bph.16389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/09/2024] [Accepted: 02/24/2024] [Indexed: 06/04/2024] Open
Abstract
G protein-coupled receptors (GPCRs) are a large family of cell surface receptors that play a critical role in nervous system function by transmitting signals between cells and their environment. They are involved in many, if not all, nervous system processes, and their dysfunction has been linked to various neurological disorders representing important drug targets. This overview emphasises the GPCRs of the nervous system, which are the research focus of the members of ERNEST COST action (CA18133) working group 'Biological roles of signal transduction'. First, the (patho)physiological role of the nervous system GPCRs in the modulation of synapse function is discussed. We then debate the (patho)physiology and pharmacology of opioid, acetylcholine, chemokine, melatonin and adhesion GPCRs in the nervous system. Finally, we address the orphan GPCRs, their implication in the nervous system function and disease, and the challenges that need to be addressed to deorphanize them.
Collapse
Affiliation(s)
- Necla Birgül Iyison
- Department of Molecular Biology and Genetics, University of Bogazici, Istanbul, Turkey
| | - Clauda Abboud
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, University of Liege, Liege, Belgium
| | - Dayana Abboud
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, University of Liege, Liege, Belgium
| | | | - Ana-Nicoleta Bondar
- Faculty of Physics, University of Bucharest, Magurele, Romania
- Forschungszentrum Jülich, Institute for Computational Biomedicine (IAS-5/INM-9), Jülich, Germany
| | - Julie Dam
- Institut Cochin, CNRS, INSERM, Université Paris Cité, Paris, France
| | - Zafiroula Georgoussi
- Laboratory of Cellular Signalling and Molecular Pharmacology, Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Jesús Giraldo
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística and Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain
- Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anemari Horvat
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| | - Christos Karoussiotis
- Laboratory of Cellular Signalling and Molecular Pharmacology, Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Alba Paz-Castro
- Molecular Pharmacology of GPCRs research group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago, Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago, Spain
| | - Miriam Scarpa
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Hannes Schihada
- Department of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Nicole Scholz
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Bilge Güvenc Tuna
- Department of Biophysics, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Nina Vardjan
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| |
Collapse
|
4
|
Li S, Luo H, Tang P, Tian C, Hu J, Lu H, Shui W. Generation of a Deep Mouse Brain Spectral Library for Transmembrane Proteome Profiling in Mental Disease Models. Mol Cell Proteomics 2024; 23:100777. [PMID: 38670310 PMCID: PMC11137342 DOI: 10.1016/j.mcpro.2024.100777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 04/28/2024] Open
Abstract
Transmembrane (TM) proteins constitute over 30% of the mammalian proteome and play essential roles in mediating cell-cell communication, synaptic transmission, and plasticity in the central nervous system. Many of these proteins, especially the G protein-coupled receptors (GPCRs), are validated or candidate drug targets for therapeutic development for mental diseases, yet their expression profiles are underrepresented in most global proteomic studies. Herein, we establish a brain TM protein-enriched spectral library based on 136 data-dependent acquisition runs acquired from various brain regions of both naïve mice and mental disease models. This spectral library comprises 3043 TM proteins including 171 GPCRs, 231 ion channels, and 598 transporters. Leveraging this library, we analyzed the data-independent acquisition data from different brain regions of two mouse models exhibiting depression- or anxiety-like behaviors. By integrating multiple informatics workflows and library sources, our study significantly expanded the mental stress-perturbed TM proteome landscape, from which a new GPCR regulator of depression was verified by in vivo pharmacological testing. In summary, we provide a high-quality mouse brain TM protein spectral library to largely increase the TM proteome coverage in specific brain regions, which would catalyze the discovery of new potential drug targets for the treatment of mental disorders.
Collapse
Affiliation(s)
- Shanshan Li
- Institutes of Biomedical Sciences and Department of Chemistry, Fudan University, Shanghai, China; iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Huoqing Luo
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China; Department of Anesthesiology & Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Pan Tang
- iHuman Institute, ShanghaiTech University, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Cuiping Tian
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Ji Hu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| | - Haojie Lu
- Institutes of Biomedical Sciences and Department of Chemistry, Fudan University, Shanghai, China.
| | - Wenqing Shui
- iHuman Institute, ShanghaiTech University, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
5
|
Anversa RG, Maddern XJ, Lawrence AJ, Walker LC. Orphan peptide and G protein-coupled receptor signalling in alcohol use disorder. Br J Pharmacol 2024; 181:595-609. [PMID: 38073127 PMCID: PMC10953447 DOI: 10.1111/bph.16301] [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: 10/14/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 01/10/2024] Open
Abstract
Neuropeptides and G protein-coupled receptors (GPCRs) have long been, and continue to be, one of the most popular target classes for drug discovery in CNS disorders, including alcohol use disorder (AUD). Yet, orphaned neuropeptide systems and receptors (oGPCR), which have no known cognate receptor or ligand, remain understudied in drug discovery and development. Orphan neuropeptides and oGPCRs are abundantly expressed within the brain and represent an unprecedented opportunity to address brain function and may hold potential as novel treatments for disease. Here, we describe the current literature regarding orphaned neuropeptides and oGPCRs implicated in AUD. Specifically, in this review, we focus on the orphaned neuropeptide cocaine- and amphetamine-regulated transcript (CART), and several oGPCRs that have been directly implicated in AUD (GPR6, GPR26, GPR88, GPR139, GPR158) and discuss their potential and pitfalls as novel treatments, and progress in identifying their cognate receptors or ligands.
Collapse
Affiliation(s)
- Roberta Goncalves Anversa
- Florey Institute of Neuroscience and Mental HealthMelbourneVICAustralia
- Florey Department of Neuroscience and Mental HealthUniversity of MelbourneMelbourneVICAustralia
| | - Xavier J. Maddern
- Florey Institute of Neuroscience and Mental HealthMelbourneVICAustralia
- Florey Department of Neuroscience and Mental HealthUniversity of MelbourneMelbourneVICAustralia
| | - Andrew J. Lawrence
- Florey Institute of Neuroscience and Mental HealthMelbourneVICAustralia
- Florey Department of Neuroscience and Mental HealthUniversity of MelbourneMelbourneVICAustralia
| | - Leigh C. Walker
- Florey Institute of Neuroscience and Mental HealthMelbourneVICAustralia
- Florey Department of Neuroscience and Mental HealthUniversity of MelbourneMelbourneVICAustralia
| |
Collapse
|
6
|
Dementieva NV, Dysin AP, Shcherbakov YS, Nikitkina EV, Musidray AA, Petrova AV, Mitrofanova OV, Plemyashov KV, Azovtseva AI, Griffin DK, Romanov MN. Risk of Sperm Disorders and Impaired Fertility in Frozen-Thawed Bull Semen: A Genome-Wide Association Study. Animals (Basel) 2024; 14:251. [PMID: 38254422 PMCID: PMC10812825 DOI: 10.3390/ani14020251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Cryopreservation is a widely used method of semen conservation in animal breeding programs. This process, however, can have a detrimental effect on sperm quality, especially in terms of its morphology. The resultant sperm disorders raise the risk of reduced sperm fertilizing ability, which poses a serious threat to the long-term efficacy of livestock reproduction and breeding. Understanding the genetic factors underlying these effects is critical for maintaining sperm quality during cryopreservation, and for animal fertility in general. In this regard, we performed a genome-wide association study to identify genomic regions associated with various cryopreservation sperm abnormalities in Holstein cattle, using single nucleotide polymorphism (SNP) markers via a high-density genotyping assay. Our analysis revealed a significant association of specific SNPs and candidate genes with absence of acrosomes, damaged cell necks and tails, as well as wrinkled acrosomes and decreased motility of cryopreserved sperm. As a result, we identified candidate genes such as POU6F2, LPCAT4, DPYD, SLC39A12 and CACNB2, as well as microRNAs (bta-mir-137 and bta-mir-2420) that may play a critical role in sperm morphology and disorders. These findings provide crucial information on the molecular mechanisms underlying acrosome integrity, motility, head abnormalities and damaged cell necks and tails of sperm after cryopreservation. Further studies with larger sample sizes, genome-wide coverage and functional validation are needed to explore causal variants in more detail, thereby elucidating the mechanisms mediating these effects. Overall, our results contribute to the understanding of genetic architecture in cryopreserved semen quality and disorders in bulls, laying the foundation for improved animal reproduction and breeding.
Collapse
Affiliation(s)
- Natalia V. Dementieva
- Russian Research Institute of Farm Animal Genetics and Breeding—Branch of the L. K. Ernst Federal Research Centre for Animal Husbandry, Pushkin, 196601 St. Petersburg, Russia; (A.P.D.); (Y.S.S.); (E.V.N.); (A.A.M.); (A.V.P.); (O.V.M.); (A.I.A.)
| | - Artem P. Dysin
- Russian Research Institute of Farm Animal Genetics and Breeding—Branch of the L. K. Ernst Federal Research Centre for Animal Husbandry, Pushkin, 196601 St. Petersburg, Russia; (A.P.D.); (Y.S.S.); (E.V.N.); (A.A.M.); (A.V.P.); (O.V.M.); (A.I.A.)
| | - Yuri S. Shcherbakov
- Russian Research Institute of Farm Animal Genetics and Breeding—Branch of the L. K. Ernst Federal Research Centre for Animal Husbandry, Pushkin, 196601 St. Petersburg, Russia; (A.P.D.); (Y.S.S.); (E.V.N.); (A.A.M.); (A.V.P.); (O.V.M.); (A.I.A.)
| | - Elena V. Nikitkina
- Russian Research Institute of Farm Animal Genetics and Breeding—Branch of the L. K. Ernst Federal Research Centre for Animal Husbandry, Pushkin, 196601 St. Petersburg, Russia; (A.P.D.); (Y.S.S.); (E.V.N.); (A.A.M.); (A.V.P.); (O.V.M.); (A.I.A.)
| | - Artem A. Musidray
- Russian Research Institute of Farm Animal Genetics and Breeding—Branch of the L. K. Ernst Federal Research Centre for Animal Husbandry, Pushkin, 196601 St. Petersburg, Russia; (A.P.D.); (Y.S.S.); (E.V.N.); (A.A.M.); (A.V.P.); (O.V.M.); (A.I.A.)
| | - Anna V. Petrova
- Russian Research Institute of Farm Animal Genetics and Breeding—Branch of the L. K. Ernst Federal Research Centre for Animal Husbandry, Pushkin, 196601 St. Petersburg, Russia; (A.P.D.); (Y.S.S.); (E.V.N.); (A.A.M.); (A.V.P.); (O.V.M.); (A.I.A.)
| | - Olga V. Mitrofanova
- Russian Research Institute of Farm Animal Genetics and Breeding—Branch of the L. K. Ernst Federal Research Centre for Animal Husbandry, Pushkin, 196601 St. Petersburg, Russia; (A.P.D.); (Y.S.S.); (E.V.N.); (A.A.M.); (A.V.P.); (O.V.M.); (A.I.A.)
| | - Kirill V. Plemyashov
- Federal State Budgetary Educational Institution of Higher Education “St. Petersburg State University of Veterinary Medicine”, 196084 St. Petersburg, Russia;
| | - Anastasiia I. Azovtseva
- Russian Research Institute of Farm Animal Genetics and Breeding—Branch of the L. K. Ernst Federal Research Centre for Animal Husbandry, Pushkin, 196601 St. Petersburg, Russia; (A.P.D.); (Y.S.S.); (E.V.N.); (A.A.M.); (A.V.P.); (O.V.M.); (A.I.A.)
| | | | - Michael N. Romanov
- School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK;
- L. K. Ernst Federal Research Centre for Animal Husbandry, Dubrovitsy, 142132 Podolsk, Moscow Oblast, Russia
| |
Collapse
|
7
|
Palamarchuk IS, Slavich GM, Vaillancourt T, Rajji TK. Stress-related cellular pathophysiology as a crosstalk risk factor for neurocognitive and psychiatric disorders. BMC Neurosci 2023; 24:65. [PMID: 38087196 PMCID: PMC10714507 DOI: 10.1186/s12868-023-00831-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/24/2023] [Indexed: 12/18/2023] Open
Abstract
In this narrative review, we examine biological processes linking psychological stress and cognition, with a focus on how psychological stress can activate multiple neurobiological mechanisms that drive cognitive decline and behavioral change. First, we describe the general neurobiology of the stress response to define neurocognitive stress reactivity. Second, we review aspects of epigenetic regulation, synaptic transmission, sex hormones, photoperiodic plasticity, and psychoneuroimmunological processes that can contribute to cognitive decline and neuropsychiatric conditions. Third, we explain mechanistic processes linking the stress response and neuropathology. Fourth, we discuss molecular nuances such as an interplay between kinases and proteins, as well as differential role of sex hormones, that can increase vulnerability to cognitive and emotional dysregulation following stress. Finally, we explicate several testable hypotheses for stress, neurocognitive, and neuropsychiatric research. Together, this work highlights how stress processes alter neurophysiology on multiple levels to increase individuals' risk for neurocognitive and psychiatric disorders, and points toward novel therapeutic targets for mitigating these effects. The resulting models can thus advance dementia and mental health research, and translational neuroscience, with an eye toward clinical application in cognitive and behavioral neurology, and psychiatry.
Collapse
Affiliation(s)
- Iryna S Palamarchuk
- Centre for Addiction and Mental Health, 1001 Queen Street West, Toronto, ON, M6J1H4, Canada.
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Sunnybrook Health Sciences Centre, Division of Neurology, Toronto, ON, Canada.
- Temerty Faculty of Medicine, Toronto Dementia Research Alliance, University of Toronto, Toronto, ON, Canada.
| | - George M Slavich
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tracy Vaillancourt
- Counselling Psychology, Faculty of Education, University of Ottawa, Ottawa, ON, Canada
- School of Psychology, Faculty of Social Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Tarek K Rajji
- Centre for Addiction and Mental Health, 1001 Queen Street West, Toronto, ON, M6J1H4, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Temerty Faculty of Medicine, Toronto Dementia Research Alliance, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
8
|
Ignatieva EV, Lashin SA, Mustafin ZS, Kolchanov NA. Evolution of human genes encoding cell surface receptors involved in the regulation of appetite: an analysis based on the phylostratigraphic age and divergence indexes. Vavilovskii Zhurnal Genet Selektsii 2023; 27:829-838. [PMID: 38213702 PMCID: PMC10777300 DOI: 10.18699/vjgb-23-96] [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: 08/11/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 01/13/2024] Open
Abstract
Genes encoding cell surface receptors make up a significant portion of the human genome (more than a thousand genes) and play an important role in gene networks. Cell surface receptors are transmembrane proteins that interact with molecules (ligands) located outside the cell. This interaction activates signal transduction pathways in the cell. A large number of exogenous ligands of various origins, including drugs, are known for cell surface receptors, which accounts for interest in them from biomedical researchers. Appetite (the desire of the animal organism to consume food) is one of the most primitive instincts that contribute to survival. However, when the supply of nutrients is stable, the mechanism of adaptation to adverse factors acquired in the course of evolution turned out to be excessive, and therefore obesity has become one of the most serious public health problems of the twenty-first century. Pathological human conditions characterized by appetite violations include both hyperphagia, which inevitably leads to obesity, and anorexia nervosa induced by psychosocial stimuli, as well as decreased appetite caused by neurodegeneration, inflammation or cancer. Understanding the evolutionary mechanisms of human diseases, especially those related to lifestyle changes that have occurred over the past 100-200 years, is of fundamental and applied importance. It is also very important to identify relationships between the evolutionary characteristics of genes in gene networks and the resistance of these networks to changes caused by mutations. The aim of the current study is to identify the distinctive features of human genes encoding cell surface receptors involved in appetite regulation using the phylostratigraphic age index (PAI) and divergence index (DI). The values of PAI and DI were analyzed for 64 human genes encoding cell surface receptors, the orthologs of which were involved in the regulation of appetite in model animal species. It turned out that the set of genes under consideration contains an increased number of genes with the same phylostratigraphic age (PAI = 5, the stage of vertebrate divergence), and almost all of these genes (28 out of 31) belong to the superfamily of G-protein coupled receptors. Apparently, the synchronized evolution of such a large group of genes (31 genes out of 64) is associated with the development of the brain as a separate organ in the first vertebrates. When studying the distribution of genes from the same set by DI values, a significant enrichment with genes having a low DIs was revealed: eight genes (GPR26, NPY1R, GHSR, ADIPOR1, DRD1, NPY2R, GPR171, NPBWR1) had extremely low DIs (less than 0.05). Such low DI values indicate that most likely these genes are subjected to stabilizing selection. It was also found that the group of genes with low DIs was enriched with genes that had brain-specific patterns of expression. In particular, GPR26, which had the lowest DI, is in the group of brain-specific genes. Because the endogenous ligand for the GPR26 receptor has not yet been identified, this gene seems to be an extremely interesting object for further theoretical and experimental research. We believe that the features of the genes encoding cell surface receptors we have identified using the evolutionary metrics PAI and DI can be a starting point for further evolutionary analysis of the gene network regulating appetite.
Collapse
Affiliation(s)
- E V Ignatieva
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - S A Lashin
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Z S Mustafin
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - N A Kolchanov
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
9
|
Wong TS, Li G, Li S, Gao W, Chen G, Gan S, Zhang M, Li H, Wu S, Du Y. G protein-coupled receptors in neurodegenerative diseases and psychiatric disorders. Signal Transduct Target Ther 2023; 8:177. [PMID: 37137892 PMCID: PMC10154768 DOI: 10.1038/s41392-023-01427-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 02/17/2023] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Neuropsychiatric disorders are multifactorial disorders with diverse aetiological factors. Identifying treatment targets is challenging because the diseases are resulting from heterogeneous biological, genetic, and environmental factors. Nevertheless, the increasing understanding of G protein-coupled receptor (GPCR) opens a new possibility in drug discovery. Harnessing our knowledge of molecular mechanisms and structural information of GPCRs will be advantageous for developing effective drugs. This review provides an overview of the role of GPCRs in various neurodegenerative and psychiatric diseases. Besides, we highlight the emerging opportunities of novel GPCR targets and address recent progress in GPCR drug development.
Collapse
Affiliation(s)
- Thian-Sze Wong
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
- School of Medicine, Tsinghua University, 100084, Beijing, China
| | - Guangzhi Li
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, 518000, Shenzhen, Guangdong, China
| | - Shiliang Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Wei Gao
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Geng Chen
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
| | - Shiyi Gan
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
| | - Manzhan Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Honglin Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China.
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China.
| | - Song Wu
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, 518000, Shenzhen, Guangdong, China.
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, 518116, Shenzhen, Guangdong, China.
| | - Yang Du
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China.
| |
Collapse
|
10
|
Gutiérrez-Rojas RA, Aguayo-Cerón KA, Vargas-De-León C, Cabrera-Becerra SE, Almanza-Pérez JC, Huang F, Villafaña S, Romero-Nava R. Glycine Effect on the Expression Profile of Orphan Receptors GPR21, GPR26, GPR39, GPR82 and GPR6 in a Model of Inflammation in 3T3-L1 Cells. Life (Basel) 2022; 12:1687. [PMID: 36362842 PMCID: PMC9696036 DOI: 10.3390/life12111687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Chronic or low-grade inflammation is a process where various immune cells are recruited from the periphery into adipose tissue. This event gives rise to localised inflammation, in addition to having a close interaction with cardiometabolic pathologies where the mediation of orphan receptors is observed. The aim of this study was to analyse the participation of the orphan receptors GPR21, GPR39, GPR82 and GPR6 in a chronic inflammatory process in 3T3-L1 cells. The 3T3-L1 cells were stimulated with TNF-α (5 ng/mL) for 60 min as an inflammatory model. Gene expression was measured by RT-qPCR. RESULTS We showed that the inflammatory stimulus of TNF-α in adipocytes decreased the expression of the orphan receptors GPR21, GPR26, GPR39, GPR82 and GPR6, which are related to low-grade inflammation. CONCLUSIONS Our results suggest that GPR21 and GPR82 are modulated by glycine, it shows a possible protective role in the presence of an inflammatory environment in adipocytes, and they could be a therapeutic target to decrease the inflammation in some diseases related to low-grade inflammation such as diabetes, obesity and metabolic syndrome.
Collapse
Affiliation(s)
| | - Karla Aidee Aguayo-Cerón
- Laboratorio de Señalización Intracelular, Sección de Estudios de Posgrado, Escuela Superior de Medicina del Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | - Cruz Vargas-De-León
- Laboratorio de Señalización Intracelular, Sección de Estudios de Posgrado, Escuela Superior de Medicina del Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
- División de Investigación, Hospital Juárez de México, Ciudad de México 07760, Mexico
| | - Sandra Edith Cabrera-Becerra
- Laboratorio de Señalización Intracelular, Sección de Estudios de Posgrado, Escuela Superior de Medicina del Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | - Julio Cesar Almanza-Pérez
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa (UAM-I), Ciudad de México 09340, Mexico
| | - Fengyang Huang
- Laboratorio de Investigación en Farmacología, Hospital Infantil de México Federico Gómez (HIMFG), Ciudad de México 06720, Mexico
| | - Santiago Villafaña
- Laboratorio de Señalización Intracelular, Sección de Estudios de Posgrado, Escuela Superior de Medicina del Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | - Rodrigo Romero-Nava
- Laboratorio de Señalización Intracelular, Sección de Estudios de Posgrado, Escuela Superior de Medicina del Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| |
Collapse
|
11
|
Ma X, Guo J, Fu Y, Shen C, Jiang P, Zhang Y, Zhang L, Yu Y, Fan J, Chai R. G protein-coupled receptors in cochlea: Potential therapeutic targets for hearing loss. Front Mol Neurosci 2022; 15:1028125. [PMID: 36311029 PMCID: PMC9596917 DOI: 10.3389/fnmol.2022.1028125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 09/21/2022] [Indexed: 11/29/2022] Open
Abstract
The prevalence of hearing loss-related diseases caused by different factors is increasing worldwide year by year. Currently, however, the patient’s hearing loss has not been effectively improved. Therefore, there is an urgent need to adopt new treatment measures and treatment techniques to help improve the therapeutic effect of hearing loss. G protein-coupled receptors (GPCRs), as crucial cell surface receptors, can widely participate in different physiological and pathological processes, particularly play an essential role in many disease occurrences and be served as promising therapeutic targets. However, no specific drugs on the market have been found to target the GPCRs of the cochlea. Interestingly, many recent studies have demonstrated that GPCRs can participate in various pathogenic process related to hearing loss in the cochlea including heredity, noise, ototoxic drugs, cochlear structure, and so on. In this review, we comprehensively summarize the functions of 53 GPCRs known in the cochlea and their relationships with hearing loss, and highlight the recent advances of new techniques used in cochlear study including cryo-EM, AI, GPCR drug screening, gene therapy vectors, and CRISPR editing technology, as well as discuss in depth the future direction of novel GPCR-based drug development and gene therapy for cochlear hearing loss. Collectively, this review is to facilitate basic and (pre-) clinical research in this area, and provide beneficial help for emerging GPCR-based cochlear therapies.
Collapse
Affiliation(s)
- Xiangyu Ma
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Jiamin Guo
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Yaoyang Fu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cangsong Shen
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Pei Jiang
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Yuan Zhang
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Research Institute of Otolaryngology, Nanjing, China
| | - Lei Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Hospital of Anhui Medical University, Hefei, China
| | - Yafeng Yu
- First Affiliated Hospital of Soochow University, Soochow, China
- *Correspondence: Yafeng Yu,
| | - Jiangang Fan
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Jiangang Fan,
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, China
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
- Renjie Chai,
| |
Collapse
|
12
|
Zhang XH, Shen CL, Wang XY, Xiong WF, Shang X, Tang LY, Zhang HX, Wan YH, Wu YB, Fei J, Yi QZ, Wang ZG. Increased Anxiety-like Behaviors in Adhesion G protein-coupled receptor A1 -/- Male But Not Female Mice are Attributable to Elevated Neuron Dendritic Density, Upregulated Postsynaptic Density Protein 95 Expression, and Abnormal Activation of the Phosphatidylinositol 3 Kinase/Protein Kinase B/Glycogen Synthase Kinase-3 and Methyl Ethyl Ketone/Extracellular Signal Regulated Kinase Pathways. Neuroscience 2022; 503:131-145. [PMID: 36115515 DOI: 10.1016/j.neuroscience.2022.09.003] [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: 05/14/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 11/19/2022]
Abstract
Adhesion G protein-coupled receptor A1 (ADGRA1) belongs to the G protein-coupled receptor (GPCR) family, and its physiological function remains largely unknown. We found that Adgra1 is highly and exclusively expressed in the brain, suggesting that Adgra1 may be involved in the regulation of neurological behaviors including anxiety, depression, learning and memory. To this end, we comprehensively analyzed the potential role of ADGRA1 in the neurobehaviors of mice by comparing Adgra1-/- and their wild-type (wt) littermates. We found that Adgra1-/- male but not female mice exhibited elevated anxiety levels in the open field, elevated plus maze, and light-dark box tests, with normal depression levels in the tail-suspension and forced-swim tests, and comparable learning and memory abilities in the Morris water maze, Y maze, fear condition, and step-down avoidance tests. Further studies showed that ADGRA1 deficiency resulted in higher dendritic branching complexity and spine density as evidenced by elevated expression levels of SYN and PSD95 in amygdalae-of male mice. Finally, we found that PI3K/AKT/GSK-3β and MEK/ERK in amygdalae of Adgra1-deficient male mice were aberrantly activated when compared to wt male mice. Together, our findings reveal an important suppressive role of ADGRA1 in anxiety control and synaptic function by regulating the PI3K/AKT/GSK-3β and MEK/ERK pathways in amygdalae of male mice, implicating a potential, therapeutic application in novel anti-anxiety drug development.
Collapse
Affiliation(s)
- Xiao-Hong Zhang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200025, China.
| | - Chun-Ling Shen
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200025, China.
| | - Xi-Yi Wang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200025, China; Department of Obstetrics and Gynecology, Tang-Du Hospital Affiliated to the Fourth Military Medical University, Xi'an 710038, China.
| | - Wen-Feng Xiong
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200025, China.
| | - Xuan Shang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200025, China.
| | - Ling-Yun Tang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200025, China.
| | - Hong-Xin Zhang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200025, China.
| | - Ying-Han Wan
- Shanghai Engineering and Technology Research Center for Model Animals, Shanghai Model Organisms Center, Inc., Shanghai 201318, China.
| | - You-Bing Wu
- Shanghai Engineering and Technology Research Center for Model Animals, Shanghai Model Organisms Center, Inc., Shanghai 201318, China.
| | - Jian Fei
- Shanghai Engineering and Technology Research Center for Model Animals, Shanghai Model Organisms Center, Inc., Shanghai 201318, China.
| | - Qi-Zhong Yi
- Psychological Medical Center, The First Hospital affiliated to Xin Jiang Medical University, Urumqi 830054, China; Xin Jiang Clinical Research Center for Mental Health, Urumqi 830054, China.
| | - Zhu-Gang Wang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200025, China; Shanghai Engineering and Technology Research Center for Model Animals, Shanghai Model Organisms Center, Inc., Shanghai 201318, China.
| |
Collapse
|
13
|
Exercise on Striatal Dopamine Level and Anxiety-Like Behavior in Male Rats after 2-VO Cerebral Ischemia. Behav Neurol 2022; 2022:2243717. [PMID: 36147220 PMCID: PMC9489419 DOI: 10.1155/2022/2243717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/20/2022] [Indexed: 11/17/2022] Open
Abstract
The purpose of this study was to discuss the effect of voluntary wheel running on striatal dopamine levels and anxiety-like behavior in rats with global cerebral ischemia. The male Sprague-Dawley rats were signed on in this study and randomly divided into following 4 groups: Control group (C group), Sham group (S group), ischemia group (I group), and 3 weeks physical exercise before ischemia group (3RI group). The rats in the 3RI group were placed in a voluntary running wheel for three weeks to exercise. Then, the rats in I and 3RI groups received bilateral carotid artery ligation (2-VO) operation. The C and S group did not perform voluntary running exercise and the bilateral common carotid arteries of S group were exposed without ligation. In vivo microdialysis was used in conjunction with high performance liquid chromatography (HPLC) and electrochemical detection to ascertain the level of dopamine in the striatum. Elevated plus maze (EPM) and open field (OF) were used to test anxiety status at 24 hours and 7days after 2-VO cerebral ischemia. Meanwhile, gait and motor coordination evaluations were carried out to eliminate the influence of non-specific motor problems. The results indicated that cerebral ischemia instigate the increase of striatal dopamine in I group rats during acute cerebral ischemia. A 3-week voluntary wheel running significantly enhances the striatal dopamine before ischemia and obstructs a further increase of dopamine during acute cerebral ischemia in 3RI group rats. At 24 hours after ischemia, striatal dopamine returned to pre-ischemic levels in 3RI group. Striatal dopamine in I group were less than pre-ischemic levels at 7 days. Behavioral data indicated that 3-week voluntary wheel running promoted recovery of anxiety-like behavior and gait were not affected by 2-VO cerebral ischemia at 24 hours post-ischemia rats. Therefore, it can be concluded that 3-week physical exercise significantly increased the striatal dopamine and improved anxiety-like behavior by inhibiting the increase of dopamine during acute cerebral ischemia and suppressing the decrease of dopamine after 24 hours and 7 days cerebral ischemia.
Collapse
|
14
|
Escobar-Ramos A, Gómez-Rivera A, Lobato-García CE, Zamilpa A, Ble-González EA, González-Cortazar M, Gallegos-García AJ, Herrera-Ruiz M. Anxiolytic effect of the heartwood of Haematoxylum campechianum L. and sappanchalcone in an in vivo model in mice. JOURNAL OF ETHNOPHARMACOLOGY 2022; 284:114764. [PMID: 34687835 DOI: 10.1016/j.jep.2021.114764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/28/2021] [Accepted: 10/19/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Haematoxylum campechianum L., is a well-known plant in the southeast region of Mexico, where it is named as "palo tinto" or "palo de Campeche", in English there are vernacular names such as "redwood", "bloodwood tree" or "campeachy wood". Traditional medicine refers its use for the treatment of different disorders including depression. AIM OF THE STUDY Considering the traditional use of this plant for the alleviation of depression, the aim of this study was the evaluation of the anxiolytic effect of the methanolic and hydroalcoholic extracts from the heartwood of Haematoxylum campechianum L., and the sappanchalchone (Sapp). Additionally, it is presented the characterization of the new compound 4-hydroxyhematoxylol (2) isolated from the hydroalcoholic extract. MATERIAL AND METHODS The anxiolytic effect of the extracts and Sapp was evaluated by using the Elevated Plus Maze (EPM) additionally the sedative effect was assessed with the Open Field Test (OFT). The chemical characterization of Sapp and 2 was performing by 1D and 2D NMR experiments. RESULTS The EPM test showed that the administration of the plant extracts increased the percentage of time spent in open arms (76.32 ± 6.35 and 66.68 ± 20.64%, respectively for the methanolic and hydroalcoholic extracts), whereas the administration of Sapp increased the percentage of time spent in open arms by 60.07 ± 14.28%, these results are similar to Diazepam (DZP, positive control) which caused an increment of 74.06 ± 23.42%. For the OFT, all of the doses evaluated for both extracts and Sapp diminished the number of rearing (R) and total corssing (TC) behavior in a similar way to the positive control (DZO) and statistically different with respect to the vehicle. CONCLUSION The results obtained showed that the polar extracts from the heartwood of Haematoxylum campechianum L. possess both anxiolytic and sedative effect and that the chalcone-type compound Sapp, isolated from the methanolic extract, is partially responsible of these activities.
Collapse
Affiliation(s)
- Armando Escobar-Ramos
- División Académica de Ciencias Básicas, Universidad Juárez Autónoma de Tabasco, Carretera Cunduacán-Jalpa Km. 0.5, Cunduacán Tabasco, 86690, Mexico
| | - Abraham Gómez-Rivera
- División Académica de Ciencias Básicas, Universidad Juárez Autónoma de Tabasco, Carretera Cunduacán-Jalpa Km. 0.5, Cunduacán Tabasco, 86690, Mexico.
| | - Carlos Ernesto Lobato-García
- División Académica de Ciencias Básicas, Universidad Juárez Autónoma de Tabasco, Carretera Cunduacán-Jalpa Km. 0.5, Cunduacán Tabasco, 86690, Mexico
| | - Alejandro Zamilpa
- Centro de Investigación Biomédica del Sur- Instituto Mexicano del Seguro Social (IMSS), Argentina No 1, Col. Centro, 62790, Xochitepec Morelos, Mexico
| | - Ever A Ble-González
- División Académica de Ciencias Básicas, Universidad Juárez Autónoma de Tabasco, Carretera Cunduacán-Jalpa Km. 0.5, Cunduacán Tabasco, 86690, Mexico
| | - Manasés González-Cortazar
- Centro de Investigación Biomédica del Sur- Instituto Mexicano del Seguro Social (IMSS), Argentina No 1, Col. Centro, 62790, Xochitepec Morelos, Mexico
| | - Ammy Joana Gallegos-García
- División Académica de Ciencias Básicas, Universidad Juárez Autónoma de Tabasco, Carretera Cunduacán-Jalpa Km. 0.5, Cunduacán Tabasco, 86690, Mexico
| | - Maribel Herrera-Ruiz
- Centro de Investigación Biomédica del Sur- Instituto Mexicano del Seguro Social (IMSS), Argentina No 1, Col. Centro, 62790, Xochitepec Morelos, Mexico.
| |
Collapse
|
15
|
Jarończyk M, Walory J. Novel Molecular Targets of Antidepressants. Molecules 2022; 27:533. [PMID: 35056845 PMCID: PMC8778443 DOI: 10.3390/molecules27020533] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 12/12/2022] Open
Abstract
Antidepressants target a variety of proteins in the central nervous system (CNS), the most important belonging to the family of G-protein coupled receptors and the family of neurotransmitter transporters. The increasing number of crystallographic structures of these proteins have significantly contributed to the knowledge of their mechanism of action, as well as to the design of new drugs. Several computational approaches such as molecular docking, molecular dynamics, and virtual screening are useful for elucidating the mechanism of drug action and are important for drug design. This review is a survey of molecular targets for antidepressants in the CNS and computer based strategies to discover novel compounds with antidepressant activity.
Collapse
|
16
|
Mantas I, Saarinen M, Xu ZQD, Svenningsson P. Update on GPCR-based targets for the development of novel antidepressants. Mol Psychiatry 2022; 27:534-558. [PMID: 33589739 PMCID: PMC8960420 DOI: 10.1038/s41380-021-01040-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 01/31/2023]
Abstract
Traditional antidepressants largely interfere with monoaminergic transport or degradation systems, taking several weeks to have their therapeutic actions. Moreover, a large proportion of depressed patients are resistant to these therapies. Several atypical antidepressants have been developed which interact with G protein coupled receptors (GPCRs) instead, as direct targeting of receptors may achieve more efficacious and faster antidepressant actions. The focus of this review is to provide an update on how distinct GPCRs mediate antidepressant actions and discuss recent insights into how GPCRs regulate the pathophysiology of Major Depressive Disorder (MDD). We also discuss the therapeutic potential of novel GPCR targets, which are appealing due to their ligand selectivity, expression pattern, or pharmacological profiles. Finally, we highlight recent advances in understanding GPCR pharmacology and structure, and how they may provide new avenues for drug development.
Collapse
Affiliation(s)
- Ioannis Mantas
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Marcus Saarinen
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Zhi-Qing David Xu
- Department of Neurobiology, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
17
|
Fan Y, Zhang L, Kong X, Liu K, Wu H. Different Exercise Time on 5-HT and Anxiety-like Behavior in the Rat With Vascular Dementia. Am J Alzheimers Dis Other Demen 2022; 37:15333175221082743. [PMID: 35344444 PMCID: PMC10581105 DOI: 10.1177/15333175221082743] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Previous studies have demonstrated that pre-exercise suppresses anxiety-like behavior, but the effects of different exercise times on vascular dementia induced anxiety-like behavior have not been well investigated. OBJECTIVE The present study aims to investigate the underlying neurochemical mechanism of different pre-vascular-dementia exercise times on 5-HT and anxiety-like behavior in rats with vascular dementia. METHODS 32 Sprague-Dawley (SD) rats were randomly divided into 4 groups: sham group (S group, n = 8), vascular dementia group (VD group, n = 8), 1-week physical exercise and vascular dementia group (1WVD group, n = 8), and 4 weeks physical exercise and vascular dementia group (4WVD group, n = 8). 1 week and 4 weeks of voluntary wheel running were used as pre-exercise training. The vascular dementia model was established by bilateral common carotid arteries occlusion (BCCAo) for 1 week. But bilateral common carotid arteries were not ligated in the sham group. The level of hippocampal 5-HT was detected with in vivo microdialysis coupled with high-performance liquid chromatography (MD-HPLC). Elevated plus maze (EPM), open field (OF), and light/dark box test were used to test anxiety-like behavior. RESULTS Compared with the C group, the hippocampal 5-HT was significantly decreased in the VD group after 1 week of ligated operation. The hippocampal 5-HT levels in 1WVD and 4WVD groups were substantially higher than the level in the VD group. The hippocampal 5-HT level has no significant difference among C, 1WVD, and 4WVD. Behavioral data suggested that the rats in the VD group developed obvious anxiety-like behavior after 1 week of ligation surgery. Still, the rats in 1WVD and 4WVD groups did not show significant anxiety-like behavior. CONCLUSION Both 1 week and 4 weeks of voluntary running wheel exercise can inhibit the anxiety-like behavior in rats with vascular dementia by upregulating 5-HT levels in the hippocampus in the VD model.
Collapse
Affiliation(s)
- Yongzhao Fan
- Capital University of Physical Education and Sports, Comprehensive Key Laboratory of Sports Ability Evaluation and Research of the General Administration of Sport of China, Beijing Key Laboratory of Sports Function Assessment and Technical Analysis, Beijing, 100191, China
| | - Linlin Zhang
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, China
| | - Xiaoyang Kong
- Capital University of Physical Education and Sports, Comprehensive Key Laboratory of Sports Ability Evaluation and Research of the General Administration of Sport of China, Beijing Key Laboratory of Sports Function Assessment and Technical Analysis, Beijing, 100191, China
| | - Kun Liu
- Capital University of Physical Education and Sports, Comprehensive Key Laboratory of Sports Ability Evaluation and Research of the General Administration of Sport of China, Beijing Key Laboratory of Sports Function Assessment and Technical Analysis, Beijing, 100191, China
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Brain Peace Science Foundation, New Haven, CT, USA
| | - Hao Wu
- Capital University of Physical Education and Sports, Comprehensive Key Laboratory of Sports Ability Evaluation and Research of the General Administration of Sport of China, Beijing Key Laboratory of Sports Function Assessment and Technical Analysis, Beijing, 100191, China
| |
Collapse
|
18
|
Roy N, Parhar I. Habenula orphan G-protein coupled receptors in the pathophysiology of fear and anxiety. Neurosci Biobehav Rev 2021; 132:870-883. [PMID: 34801259 DOI: 10.1016/j.neubiorev.2021.11.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/02/2021] [Accepted: 11/08/2021] [Indexed: 10/19/2022]
Abstract
The phasic emotion, fear, and the tonic emotion, anxiety, have been conventionally inspected in clinical frameworks to epitomize memory acquisition, storage, and retrieval. However, inappropriate expression of learned fear in a safe environment and its resistance to suppression is a cardinal feature of various fear-related disorders. A significant body of literature suggests the involvement of extra-amygdala circuitry in fear disorders. Consistent with this view, the present review underlies incentives for the association between the habenula and fear memory. G protein-coupled receptors (GPCRs) are important to understand the molecular mechanisms central to fear learning due to their neuromodulatory role. The efficacy of a pharmacological strategy aimed at exploiting habenular-GPCR desensitization machinery can serve as a therapeutic target combating the pathophysiology of fear disorders. Originating from this milieu, the conserved nature of orphan GPCRs in the brain, with some having the highest expression in the habenula can lead to recent endeavors in understanding its functionality in fear circuitry.
Collapse
Affiliation(s)
- Nisa Roy
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia.
| | - Ishwar Parhar
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
19
|
Monteiro FA, Miranda RM, Samina MC, Dias AF, Raposo AASF, Oliveira P, Reguenga C, Castro DS, Lima D. Tlx3 Exerts Direct Control in Specifying Excitatory Over Inhibitory Neurons in the Dorsal Spinal Cord. Front Cell Dev Biol 2021; 9:642697. [PMID: 33996801 PMCID: PMC8117147 DOI: 10.3389/fcell.2021.642697] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/30/2021] [Indexed: 11/28/2022] Open
Abstract
The spinal cord dorsal horn is a major station for integration and relay of somatosensory information and comprises both excitatory and inhibitory neuronal populations. The homeobox gene Tlx3 acts as a selector gene to control the development of late-born excitatory (dILB) neurons by specifying glutamatergic transmitter fate in dorsal spinal cord. However, since Tlx3 direct transcriptional targets remain largely unknown, it remains to be uncovered how Tlx3 functions to promote excitatory cell fate. Here we combined a genomics approach based on chromatin immunoprecipitation followed by next generation sequencing (ChIP-seq) and expression profiling, with validation experiments in Tlx3 null embryos, to characterize the transcriptional program of Tlx3 in mouse embryonic dorsal spinal cord. We found most dILB neuron specific genes previously identified to be directly activated by Tlx3. Surprisingly, we found Tlx3 also directly represses many genes associated with the alternative inhibitory dILA neuronal fate. In both cases, direct targets include transcription factors and terminal differentiation genes, showing that Tlx3 directly controls cell identity at distinct levels. Our findings provide a molecular frame for the master regulatory role of Tlx3 in developing glutamatergic dILB neurons. In addition, they suggest a novel function for Tlx3 as direct repressor of GABAergic dILA identity, pointing to how generation of the two alternative cell fates being tightly coupled.
Collapse
Affiliation(s)
- Filipe A Monteiro
- Unidade de Biologia Experimental, Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,Pain Research Group, Instituto de Biologia Molecular e Celular, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Rafael M Miranda
- Unidade de Biologia Experimental, Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,Pain Research Group, Instituto de Biologia Molecular e Celular, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Marta C Samina
- Unidade de Biologia Experimental, Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,Pain Research Group, Instituto de Biologia Molecular e Celular, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Ana F Dias
- Pain Research Group, Instituto de Biologia Molecular e Celular, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Alexandre A S F Raposo
- Molecular Neurobiology Group, Instituto Gulbenkian de Ciência, Oeiras, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Patrícia Oliveira
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Diagnostics, Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Carlos Reguenga
- Unidade de Biologia Experimental, Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,Pain Research Group, Instituto de Biologia Molecular e Celular, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Diogo S Castro
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Molecular Neurobiology Group, Instituto Gulbenkian de Ciência, Oeiras, Portugal.,Stem Cells & Neurogenesis Group, Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Deolinda Lima
- Unidade de Biologia Experimental, Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,Pain Research Group, Instituto de Biologia Molecular e Celular, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| |
Collapse
|
20
|
Liu F, Yang W, Hu M, Zhang Y, Sun B, Yang H, Brosius J, Deng C. Constitutive activity of GPR26 regulated by ubiquitin-dependent degradation and its antitumor role. FEBS J 2021; 288:4655-4682. [PMID: 33577134 DOI: 10.1111/febs.15763] [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: 10/15/2020] [Revised: 01/13/2021] [Accepted: 02/11/2021] [Indexed: 02/05/2023]
Abstract
G protein-coupled receptors (GPCRs) play important roles in many physiological functions and numerous diseases. In addition to the classic ligand-stimulated receptor activity, an increasing number of studies have established that many GPCRs function constitutively in a receptor dose-dependent manner. Previous observations showed that following gene transfection, little or no protein was detectable for certain GPCRs (designated apparent state A), such as GPR26, GPR39, GPR78, GPR133, GPR139, BRS3, and LGR5, which showed strong constitutive activities. When we lysed cells in the immediate presence of western blot loading buffer, a significant increase of protein levels was detected (actual state B), which was much closer to the true expression levels under physiological conditions. GPR26 was chosen for further functional experiments as the actual state B. We identified an important ubiquitination site, K286, as well as the ubiquitin ligase E3 homologous to the E6-associated protein carboxyl terminus domain containing 3 interacting with GPR26. The pronounced differences in the protein expression and constitutive activity of GPR26 were a consequence of the ubiquitin-mediated rapid degradation mechanism. Furthermore, we identified in vitro and in vivo antitumor activity associated with high expression levels and constitutive activity of GPR26 in liver cancer cells. Hence, GPR26 could act as an antitumor gene for hepatocellular carcinoma. This study also represents the actual state B of a batch of GPCRs that actually play potentially important roles in physiological functions by their constitutive activity, which is controlled by rapid ubiquitin-dependent degradation.
Collapse
Affiliation(s)
- Fang Liu
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, China
| | - Wei Yang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, China
| | - Minghui Hu
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, China
| | - Yong Zhang
- West China - Washington Mitochondria and Metabolism Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Beicheng Sun
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, China
| | - Hao Yang
- Key Laboratory of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University, Chengdu, China
| | - Juergen Brosius
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.,Institute of Experimental Pathology, ZMBE, University of Münster, Germany
| | - Cheng Deng
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, China
| |
Collapse
|
21
|
Tiwari P, Fanibunda SE, Kapri D, Vasaya S, Pati S, Vaidya VA. GPCR signaling: role in mediating the effects of early adversity in psychiatric disorders. FEBS J 2021; 288:2602-2621. [DOI: 10.1111/febs.15738] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/11/2021] [Accepted: 01/27/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Praachi Tiwari
- Department of Biological Sciences Tata Institute of Fundamental Research Mumbai India
| | - Sashaina E. Fanibunda
- Department of Biological Sciences Tata Institute of Fundamental Research Mumbai India
- Medical Research Centre Kasturba Health Society Mumbai India
| | - Darshana Kapri
- Department of Biological Sciences Tata Institute of Fundamental Research Mumbai India
| | - Shweta Vasaya
- Department of Biological Sciences Tata Institute of Fundamental Research Mumbai India
| | - Sthitapranjya Pati
- Department of Biological Sciences Tata Institute of Fundamental Research Mumbai India
| | - Vidita A. Vaidya
- Department of Biological Sciences Tata Institute of Fundamental Research Mumbai India
| |
Collapse
|
22
|
Watkins LR, Orlandi C. Orphan G Protein Coupled Receptors in Affective Disorders. Genes (Basel) 2020; 11:E694. [PMID: 32599826 PMCID: PMC7349732 DOI: 10.3390/genes11060694] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/20/2020] [Accepted: 06/21/2020] [Indexed: 12/12/2022] Open
Abstract
G protein coupled receptors (GPCRs) are the main mediators of signal transduction in the central nervous system. Therefore, it is not surprising that many GPCRs have long been investigated for their role in the development of anxiety and mood disorders, as well as in the mechanism of action of antidepressant therapies. Importantly, the endogenous ligands for a large group of GPCRs have not yet been identified and are therefore known as orphan GPCRs (oGPCRs). Nonetheless, growing evidence from animal studies, together with genome wide association studies (GWAS) and post-mortem transcriptomic analysis in patients, pointed at many oGPCRs as potential pharmacological targets. Among these discoveries, we summarize in this review how emotional behaviors are modulated by the following oGPCRs: ADGRB2 (BAI2), ADGRG1 (GPR56), GPR3, GPR26, GPR37, GPR50, GPR52, GPR61, GPR62, GPR88, GPR135, GPR158, and GPRC5B.
Collapse
Affiliation(s)
| | - Cesare Orlandi
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA;
| |
Collapse
|
23
|
Neasta J, Darcq E, Jeanblanc J, Carnicella S, Ben Hamida S. GPCR and Alcohol-Related Behaviors in Genetically Modified Mice. Neurotherapeutics 2020; 17:17-42. [PMID: 31919661 PMCID: PMC7007453 DOI: 10.1007/s13311-019-00828-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
G protein-coupled receptors (GPCRs) constitute the largest class of cell surface signaling receptors and regulate major neurobiological processes. Accordingly, GPCRs represent primary targets for the treatment of brain disorders. Several human genetic polymorphisms affecting GPCRs have been associated to different components of alcohol use disorder (AUD). Moreover, GPCRs have been reported to contribute to several features of alcohol-related behaviors in animal models. Besides traditional pharmacological tools, genetic-based approaches mostly aimed at deleting GPCR genes provided substantial information on how key GPCRs drive alcohol-related behaviors. In this review, we summarize the alcohol phenotypes that ensue from genetic manipulation, in particular gene deletion, of key GPCRs in rodents. We focused on GPCRs that belong to fundamental neuronal systems that have been shown as potential targets for the development of AUD treatment. Data are reviewed with particular emphasis on alcohol reward, seeking, and consumption which are behaviors that capture essential aspects of AUD. Literature survey indicates that in most cases, there is still a gap in defining the intracellular transducers and the functional crosstalk of GPCRs as well as the neuronal populations in which their signaling regulates alcohol actions. Further, the implication of only a few orphan GPCRs has been so far investigated in animal models. Combining advanced pharmacological technologies with more specific genetically modified animals and behavioral preclinical models is likely necessary to deepen our understanding in how GPCR signaling contributes to AUD and for drug discovery.
Collapse
Affiliation(s)
- Jérémie Neasta
- Laboratoire de Pharmacologie, Faculté de Pharmacie, University of Montpellier, 34093, Montpellier, France
| | - Emmanuel Darcq
- Douglas Hospital Research Center, Department of Psychiatry, McGill University, 6875 Boulevard LaSalle, Montreal, Quebec, H4H 1R3, Canada
| | - Jérôme Jeanblanc
- Research Group on Alcohol and Pharmacodependences-INSERM U1247, University of Picardie Jules Verne, 80025, Amiens, France
| | - Sebastien Carnicella
- INSERM U1216, Grenoble Institut des Neurosciences (GIN), University of Grenoble Alpes, 38000, Grenoble, France
| | - Sami Ben Hamida
- Douglas Hospital Research Center, Department of Psychiatry, McGill University, 6875 Boulevard LaSalle, Montreal, Quebec, H4H 1R3, Canada.
| |
Collapse
|
24
|
Yu Y, Xu D, Cheng S, Zhang L, Shi Z, Qin J, Zhang Z, Wang H. Prenatal ethanol exposure enhances the susceptibility to depressive behavior of adult offspring rats fed a high‑fat diet by affecting BDNF‑associated pathway. Int J Mol Med 2019; 45:365-374. [PMID: 31894308 PMCID: PMC6984802 DOI: 10.3892/ijmm.2019.4436] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 11/05/2019] [Indexed: 12/27/2022] Open
Abstract
Epidemiological studies have shown that exposure to ethanol during pregnancy can result in an increased risk for depression in offspring. A 'brain-derived neurotrophic factor (BDNF) hypothesis' has been proposed to help explain the pathogenic mechanism of depression. This study was designed to verify the enhanced susceptibility to depression in prenatal ethanol exposure (PEE) offspring rats and explore possible intrauterine programming mechanisms related to the BDNF signaling pathway. Pregnant rats were intragastrically administrated ethanol (4 g/kg/day) from gestational day 11 until term delivery. All offspring rats were given a high-fat diet after weaning. Then the behavior tests, including sucrose preference test and open field test, were performed to adult offspring rats. The histomorphology of hippocampus was examined, and the expression of genes related to the BDNF signaling pathway was detected in the hippocampus of PEE offspring. The PEE female adult offspring rats showed depression-like behavior, with obvious morphological injury in hippocampus. Additionally, the mRNA expression levels of glucocorticoid receptor (GR) and BDNF pathway-associated genes were changed in hippocampus. Multigene RT-qPCR also revealed that the mRNA expression levels for BDNF pathway-associated genes and synaptic plasticity genes were decreased in the hippocampus of fetal offspring rats in the PEE group. The underlying mechanism involves an increased GR expression that constantly suppresses the BDNF signaling pathway, and aggravates the functional insult to the hippo-campus, resulting in an increased susceptibility to depression among PEE female adult offspring rats. Results of the present study provide theoretical and experimental evidence that can be used for the early prevention and treatment of depression.
Collapse
Affiliation(s)
- Ying Yu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei 430071, P.R. China
| | - Dan Xu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei 430071, P.R. China
| | - Siyuan Cheng
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei 430071, P.R. China
| | - Li Zhang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei 430071, P.R. China
| | - Zhaokun Shi
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jun Qin
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zhaohui Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
25
|
Xu W, Morford J, Mauvais-Jarvis F. Emerging role of testosterone in pancreatic β-cell function and insulin secretion. J Endocrinol 2019; 240:JOE-18-0573.R1. [PMID: 30601759 PMCID: PMC6602868 DOI: 10.1530/joe-18-0573] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 12/24/2018] [Indexed: 12/16/2022]
Abstract
One of the most sexually dimorphic aspects of metabolic regulation is the bidirectional modulation of glucose homeostasis by testosterone in male and females. Severe testosterone deficiency predisposes men to type 2 diabetes (T2D), while in contrast, androgen excess predisposes women to hyperglycemia. The role of androgen deficiency and excess in promoting visceral obesity and insulin resistance in men and women respectively is well established. However, although it is established that hyperglycemia requires β cell dysfunction to develop, the role of testosterone in β cell function is less understood. This review discusses recent evidence that the androgen receptor (AR) is present in male and female β cells. In males, testosterone action on AR in β cells enhances glucose-stimulated insulin secretion by potentiating the insulinotropic action of glucagon-like peptide-1. In females, excess testosterone action via AR in β cells promotes insulin hypersecretion leading to oxidative injury, which in turn predisposes to T2D.
Collapse
Affiliation(s)
- Weiwei Xu
- W Xu, Division of Endocrinology and Metabolism, Tulane University, New Orleans, United States
| | - Jamie Morford
- J Morford, Division of Endocrinology and Metabolism, Tulane University, New Orleans, United States
| | - Franck Mauvais-Jarvis
- F Mauvais-Jarvis, Division of Endocrinology and Metabolism, Tulane University, New Orleans, United States
| |
Collapse
|
26
|
Summers CH, Yaeger JDW, Staton CD, Arendt DH, Summers TR. Orexin/hypocretin receptor modulation of anxiolytic and antidepressive responses during social stress and decision-making: Potential for therapy. Brain Res 2018; 1731:146085. [PMID: 30590027 DOI: 10.1016/j.brainres.2018.12.036] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 12/15/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022]
Abstract
Hypothalmic orexin/hypocretin (Orx) neurons in the lateral and dorsomedial perifornical region (LH-DMH/PeF) innervate broadly throughout the brain, and receive similar inputs. This wide distribution, as well as two Orx peptides (OrxA and OrxB) and two Orx receptors (Orx1 and Orx2) allow for functionally related but distinctive behavioral outcomes, that include arousal, sleep-wake regulation, food seeking, metabolism, feeding, reward, addiction, and learning. These are all motivational functions, and tie the orexin systems to anxiety and depression as well. We present evidence, that for affective behavior, Orx1 and Orx2 receptors appear to have opposing functions. The majority of research on anxiety- and depression-related outcomes has focused on Orx1 receptors, which appear to have primarily anxiogenic and pro-depressive actions. Although there is significant research suggesting contrary findings, the primary potential for pharmacotherapies linked to the Orx1 receptor is via antagonists to block anxious and depressive behavior. Dual orexin receptor antagonists have been approved for treatment of sleep disorders, and are likely candidates for adaptation for affect disorder treatments. However, we present evidence here that demonstrates the Orx2 receptors are anxiolytic and antidepressive. Using a new experimental pre-clinical model of anxious and depressive behavior stimulated by social stress and decision-making that produces two stable behavioral phenotypes, Escape/Resilient and Stay/Susceptible, we tested the effects of intracerebroventricular injections of Orx2 agonist and antagonist drugs. Over ten behavioral measures, we have demonstrated that Orx2 agonists promote resilience, as well as anxiolytic and antidepressive behavior. In contrast, Orx2 antagonists or knockdown kindle anxious and pro-depressive behavior plus increase susceptibility. The results suggest that the Orx2 receptor may be a useful target for pharmacotherapies to treat anxiety and depression.
Collapse
Affiliation(s)
- Cliff H Summers
- Department of Biology, University of South Dakota, Vermillion, SD 57069 USA; Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069 USA; Veterans Affairs Research Service, Sioux Falls VA Health Care System, Sioux Falls, SD 57105 USA.
| | - Jazmine D W Yaeger
- Department of Biology, University of South Dakota, Vermillion, SD 57069 USA; Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069 USA; Veterans Affairs Research Service, Sioux Falls VA Health Care System, Sioux Falls, SD 57105 USA
| | - Clarissa D Staton
- Department of Biology, University of South Dakota, Vermillion, SD 57069 USA; Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069 USA; Veterans Affairs Research Service, Sioux Falls VA Health Care System, Sioux Falls, SD 57105 USA
| | - David H Arendt
- Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069 USA
| | - Tangi R Summers
- Department of Biology, University of South Dakota, Vermillion, SD 57069 USA; Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069 USA; Veterans Affairs Research Service, Sioux Falls VA Health Care System, Sioux Falls, SD 57105 USA
| |
Collapse
|
27
|
Wang X, Shen C, Chen X, Wang J, Cui X, Wang Y, Zhang H, Tang L, Lu S, Fei J, Wang Z. Tafa-2 plays an essential role in neuronal survival and neurobiological function in mice. Acta Biochim Biophys Sin (Shanghai) 2018; 50:984-995. [PMID: 30137205 PMCID: PMC6185136 DOI: 10.1093/abbs/gmy097] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/25/2018] [Indexed: 12/26/2022] Open
Abstract
Tafa is a family of small secreted proteins with conserved cysteine residues and restricted expression in the brain. It is composed of five highly homologous genes referred to as Tafa-1 to -5. Among them, Tafa-2 is identified as one of the potential genes responsible for intellectual deficiency in a patient with mild mental retardation. To investigate the biological function of Tafa-2 in vivo, Tafa-2 knockout mice were generated. The mutant mice grew and developed normally but exhibited impairments in spatial learning and memory in Morris water maze test and impairments in short- and long-term memory in novel object recognition test, accompanied with increased level of anxiety-like behaviors in open-field test and elevated plus maze test, and decreased level of depression-like behaviors in forced-swim test and tail-suspension test. Further examinations revealed that Tafa-2 deficiency causes severe neuronal reduction and increased apoptosis in the brain of Tafa-2-/- mice via downregulation of PI3K/Akt and MAPK/Erk pathways. Conformably, the expression levels of CREB target genes including BDNF, c-fos and NF1, and CBP were found to be reduced in the brain of Tafa-2-/- mice. Taken together, our data indicate that Tafa-2 may function as a neurotrophic factor essential for neuronal survival and neurobiological functions.
Collapse
Affiliation(s)
- Xiyi Wang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Shanghai Research Center for Model Organisms, Shanghai, China
| | - Chunling Shen
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Xuejiao Chen
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Jinjin Wang
- Shanghai Research Center for Model Organisms, Shanghai, China
| | - Xiaofang Cui
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Yicheng Wang
- Shanghai Research Center for Model Organisms, Shanghai, China
| | - Hongxin Zhang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Lingyun Tang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Shunyuan Lu
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Jian Fei
- Shanghai Research Center for Model Organisms, Shanghai, China
| | - Zhugang Wang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Shanghai Research Center for Model Organisms, Shanghai, China
| |
Collapse
|
28
|
Chamessian A, Young M, Qadri Y, Berta T, Ji RR, Van de Ven T. Transcriptional Profiling of Somatostatin Interneurons in the Spinal Dorsal Horn. Sci Rep 2018; 8:6809. [PMID: 29717160 PMCID: PMC5931607 DOI: 10.1038/s41598-018-25110-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 04/13/2018] [Indexed: 01/08/2023] Open
Abstract
The spinal dorsal horn (SDH) is comprised of distinct neuronal populations that process different somatosensory modalities. Somatostatin (SST)-expressing interneurons in the SDH have been implicated specifically in mediating mechanical pain. Identifying the transcriptomic profile of SST neurons could elucidate the unique genetic features of this population and enable selective analgesic targeting. To that end, we combined the Isolation of Nuclei Tagged in Specific Cell Types (INTACT) method and Fluorescence Activated Nuclei Sorting (FANS) to capture tagged SST nuclei in the SDH of adult male mice. Using RNA-sequencing (RNA-seq), we uncovered more than 13,000 genes. Differential gene expression analysis revealed more than 900 genes with at least 2-fold enrichment. In addition to many known dorsal horn genes, we identified and validated several novel transcripts from pharmacologically tractable functional classes: Carbonic Anhydrase 12 (Car12), Phosphodiesterase 11 A (Pde11a), and Protease-Activated Receptor 3 (F2rl2). In situ hybridization of these novel genes showed differential expression patterns in the SDH, demonstrating the presence of transcriptionally distinct subpopulations within the SST population. Overall, our findings provide new insights into the gene repertoire of SST dorsal horn neurons and reveal several novel targets for pharmacological modulation of this pain-mediating population and treatment of pathological pain.
Collapse
Affiliation(s)
- Alexander Chamessian
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710, USA. .,Medical Scientist Training Program, Duke University School of Medicine, Durham, North Carolina, 27710, USA. .,Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, 27710, USA.
| | - Michael Young
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Yawar Qadri
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, 45267, USA
| | - Ru-Rong Ji
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710, USA.,Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Thomas Van de Ven
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| |
Collapse
|
29
|
Alavi MS, Shamsizadeh A, Azhdari-Zarmehri H, Roohbakhsh A. Orphan G protein-coupled receptors: The role in CNS disorders. Biomed Pharmacother 2017; 98:222-232. [PMID: 29268243 DOI: 10.1016/j.biopha.2017.12.056] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 12/12/2017] [Accepted: 12/14/2017] [Indexed: 12/20/2022] Open
Abstract
There are various types of receptors in the central nervous system (CNS). G protein-coupled receptors (GPCRs) have the highest expression with a wide range of physiological functions. A newer sub group of these receptors namely orphan GPCRs have been discovered. GPR3, GPR6, GPR17, GPR26, GPR37, GPR39, GPR40, GPR50, GPR52, GPR54, GPR55, GPR85, GPR88, GPR103, and GPR139 are the selected orphan GPCRs for this article. Their roles in the central nervous system have not been understood well so far. However, recent studies show that they may have very important functions in the CNS. Hence, in the present study, we reviewed most recent findings regarding the physiological roles of the selected orphan GPCRs in the CNS. After a brief presentation of each receptor, considering the results from genetic and pharmacological manipulation of the receptors, their roles in the pathophysiology of different diseases and disorders including anxiety, depression, schizophrenia, epilepsy, Alzheimer's disease, Parkinson's disease, and substance abuse will be discussed. At present, our knowledge regarding the role of GPCRs in the brain is very limited. However, previous limited studies show that orphan GPCRs have an important place in psychopharmacology and these receptors are potential new targets for the treatment of major CNS diseases.
Collapse
Affiliation(s)
- Mohaddeseh Sadat Alavi
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Shamsizadeh
- Physiology-Pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hassan Azhdari-Zarmehri
- Department of Basic Medical Sciences and Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Ali Roohbakhsh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
30
|
Chen X, Wang X, Tang L, Wang J, Shen C, Liu J, Lu S, Zhang H, Kuang Y, Fei J, Wang Z. Nhe5 deficiency enhances learning and memory via upregulating Bdnf/TrkB signaling in mice. Am J Med Genet B Neuropsychiatr Genet 2017; 174:828-838. [PMID: 28981195 DOI: 10.1002/ajmg.b.32600] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 07/10/2017] [Accepted: 09/05/2017] [Indexed: 11/11/2022]
Abstract
Nhe5, a Na+ /H+ exchanger, is predominantly expressed in brain tissue and is proposed to act as a negative regulator of dendritic spine growth. Up to now, its physiological function in vivo remains unclear. Here we show that Nhe5-deficient mice exhibit markedly enhanced learning and memory in Morris water maze, novel object recognition, and passive avoidance task. Meanwhile, the pre- and post-synaptic components, synaptophysin (Syn) and post-synaptic density 95 (PSD95) expression levels were found increased in hippocampal regions lacking of Nhe5, suggesting a possible alterations in neuronal synaptic structure and function in Nhe5-/- mice. Further study reveals that Nhe5 deficiency leads to higher Bdnf expression levels, followed by increased phosphorylated TrkB and PLCγ levels, indicating that Bdnf/TrkB signaling is activated due to Nhe5 deficiency. Moreover, the corresponding brain regions of Nhe5-/- mice display elevated ERK/CaMKII/CREB phosphorylation levels. Taken together, these findings uncover a novel physiological function of Nhe5 in regulating learning and memory, further implying Nhe5 as a potential therapeutic target for improving cognition.
Collapse
Affiliation(s)
- Xuejiao Chen
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine of Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China.,Shanghai Research Center for Model Organisms, Shanghai, China
| | - Xiyi Wang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine of Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China.,Shanghai Research Center for Model Organisms, Shanghai, China
| | - Lingyun Tang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine of Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Jinjin Wang
- Shanghai Research Center for Model Organisms, Shanghai, China
| | - Chunling Shen
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine of Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China.,Shanghai Research Center for Model Organisms, Shanghai, China
| | - Jianbing Liu
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine of Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China.,Shanghai Research Center for Model Organisms, Shanghai, China
| | - Shunyuan Lu
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine of Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Hongxin Zhang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine of Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Ying Kuang
- Shanghai Research Center for Model Organisms, Shanghai, China
| | - Jian Fei
- Shanghai Research Center for Model Organisms, Shanghai, China
| | - Zhugang Wang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine of Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China.,Shanghai Research Center for Model Organisms, Shanghai, China.,Department of Medical Genetics, E-Institutes of Shanghai Universities, SJTUSM, Shanghai, China
| |
Collapse
|
31
|
Neanderthal-Derived Genetic Variation Shapes Modern Human Cranium and Brain. Sci Rep 2017; 7:6308. [PMID: 28740249 PMCID: PMC5524936 DOI: 10.1038/s41598-017-06587-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 05/24/2017] [Indexed: 01/25/2023] Open
Abstract
Before their disappearance from the fossil record approximately 40,000 years ago, Neanderthals, the ancient hominin lineage most closely related to modern humans, interbred with ancestors of present-day humans. The legacy of this gene flow persists through Neanderthal-derived variants that survive in modern human DNA; however, the neural implications of this inheritance are uncertain. Here, using MRI in a large cohort of healthy individuals of European-descent, we show that the amount of Neanderthal-originating polymorphism carried in living humans is related to cranial and brain morphology. First, as a validation of our approach, we demonstrate that a greater load of Neanderthal-derived genetic variants (higher “NeanderScore”) is associated with skull shapes resembling those of known Neanderthal cranial remains, particularly in occipital and parietal bones. Next, we demonstrate convergent NeanderScore-related findings in the brain (measured by gray- and white-matter volume, sulcal depth, and gyrification index) that localize to the visual cortex and intraparietal sulcus. This work provides insights into ancestral human neurobiology and suggests that Neanderthal-derived genetic variation is neurologically functional in the contemporary population.
Collapse
|
32
|
Xu W, Niu T, Xu B, Navarro G, Schipma MJ, Mauvais-Jarvis F. Androgen receptor-deficient islet β-cells exhibit alteration in genetic markers of insulin secretion and inflammation. A transcriptome analysis in the male mouse. J Diabetes Complications 2017; 31:787-795. [PMID: 28343791 PMCID: PMC5472375 DOI: 10.1016/j.jdiacomp.2017.03.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/06/2017] [Accepted: 03/06/2017] [Indexed: 11/19/2022]
Abstract
AIMS Testosterone action is mediated via the androgen receptor (AR). We have reported that male mice lacking AR selectively in β-cells (βARKO-/y) develop decreased glucose-stimulated insulin secretion (GSIS), producing glucose intolerance. We showed that testosterone action on AR in β-cells amplifies the insulinotropic action of GLP-1 on its receptor via a cAMP-dependent protein kinase-A pathway. METHODS To investigate AR-dependent gene networks in β-cells, we performed a high throughput whole transcriptome sequencing (RNA-Seq) in islets from male βARKO-/y and control mice. RESULTS We identified 214 differentially expressed genes (DEGs) (158 up- and 56 down-regulated) with a false discovery rate (FDR) < 0.05 and a fold change (FC) > 2. Our analysis of individual transcripts revealed alterations in β-cell genes involved in cellular inflammation/stress and insulin secretion. Based on 312 DEGs with an FDR < 0.05, the pathway analysis revealed 23 significantly enriched pathways, including cytokine-cytokine receptor interaction, Jak-STAT signaling, insulin signaling, MAPK signaling, type 2 diabetes (T2D) and pancreatic secretion. The gene ontology analysis confirmed the results of the individual DEGs and the pathway analysis in showing enriched biological processes encompassing inflammation, ion transport, exocytosis and insulin secretion. CONCLUSIONS AR-deficient islets exhibit altered expression of genes involved in inflammation and insulin secretion demonstrating the importance of androgen action in β-cell health in the male with implications for T2D development in men.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Crosses, Genetic
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Gene Expression Profiling
- Gene Expression Regulation
- Gene Ontology
- Genetic Markers
- High-Throughput Nucleotide Sequencing
- Insulin/metabolism
- Insulin Secretion
- Insulin-Secreting Cells/immunology
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/pathology
- Islets of Langerhans/immunology
- Islets of Langerhans/metabolism
- Islets of Langerhans/pathology
- Male
- Mice
- Mice, Knockout
- Mice, Transgenic
- Models, Biological
- Organ Specificity
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Sequence Analysis, RNA
- Transcriptome
Collapse
Affiliation(s)
- Weiwei Xu
- Diabetes Discovery Research and Gender Medicine Laboratory, Department of Medicine, Section of Endocrinology and Metabolism, Tulane University School of Medicine, New Orleans
| | - Tianhua Niu
- Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA
| | - Beibei Xu
- Diabetes Discovery Research and Gender Medicine Laboratory, Department of Medicine, Section of Endocrinology and Metabolism, Tulane University School of Medicine, New Orleans
| | - Guadalupe Navarro
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Matthew J Schipma
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Franck Mauvais-Jarvis
- Diabetes Discovery Research and Gender Medicine Laboratory, Department of Medicine, Section of Endocrinology and Metabolism, Tulane University School of Medicine, New Orleans.
| |
Collapse
|
33
|
Khan MZ, He L. Neuro-psychopharmacological perspective of Orphan receptors of Rhodopsin (class A) family of G protein-coupled receptors. Psychopharmacology (Berl) 2017; 234:1181-1207. [PMID: 28289782 DOI: 10.1007/s00213-017-4586-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 02/27/2017] [Indexed: 12/28/2022]
Abstract
BACKGROUND In the central nervous system (CNS), G protein-coupled receptors (GPCRs) are the most fruitful targets for neuropsychopharmacological drug development. Rhodopsin (class A) is the most studied class of GPCR and includes orphan receptors for which the endogenous ligand is not known or is unclear. Characterization of orphan GPCRs has proven to be challenging, and the production pace of GPCR-based drugs has been incredibly slow. OBJECTIVE Determination of the functions of these receptors may provide unexpected insight into physiological and neuropathological processes. Advances in various methods and techniques to investigate orphan receptors including in situ hybridization and knockdown/knockout (KD/KO) showed extensive expression of these receptors in the mammalian brain and unmasked their physiological and neuropathological roles. Due to these rapid progress and development, orphan GPCRs are rising as a new and promising class of drug targets for neurodegenerative diseases and psychiatric disorders. CONCLUSION This review presents a neuropsychopharmacological perspective of 26 orphan receptors of rhodopsin (class A) family, namely GPR3, GPR6, GPR12, GPR17, GPR26, GPR35, GPR39, GPR48, GPR49, GPR50, GPR52, GPR55, GPR61, GPR62, GPR63, GPR68, GPR75, GPR78, GPR83, GPR84, GPR85, GPR88, GPR153, GPR162, GPR171, and TAAR6. We discussed the expression of these receptors in mammalian brain and their physiological roles. Furthermore, we have briefly highlighted their roles in neurodegenerative diseases and psychiatric disorders including Alzheimer's disease, Parkinson's disease, neuroinflammation, inflammatory pain, bipolar and schizophrenic disorders, epilepsy, anxiety, and depression.
Collapse
Affiliation(s)
- Muhammad Zahid Khan
- Department of Pharmacology, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing, Jiangsu Province, 210009, China.
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing, Jiangsu Province, 210009, China
| |
Collapse
|
34
|
Nedd4-2 haploinsufficiency causes hyperactivity and increased sensitivity to inflammatory stimuli. Sci Rep 2016; 6:32957. [PMID: 27604420 PMCID: PMC5015076 DOI: 10.1038/srep32957] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/16/2016] [Indexed: 01/01/2023] Open
Abstract
Nedd4-2 (NEDD4L in humans) is a ubiquitin protein ligase best known for its role in regulating ion channel internalization and turnover. Nedd4-2 deletion in mice causes perinatal lethality associated with increased epithelial sodium channel (ENaC) expression in lung and kidney. Abundant data suggest that Nedd4-2 plays a role in neuronal functions and may be linked to epilepsy and dyslexia in humans. We used a mouse model of Nedd4-2 haploinsufficiency to investigate whether an alteration in Nedd4-2 levels of expression affects general nervous system functions. We found that Nedd4-2 heterozygous mice are hyperactive, have increased basal synaptic transmission and have enhanced sensitivity to inflammatory pain. Thus, Nedd4-2 heterozygous mice provide a new genetic model to study inflammatory pain. These data also suggest that in human, SNPs affecting NEDD4L levels may be involved in the development of neuropsychological deficits and peripheral neuropathies and may help unveil the genetic basis of comorbidities.
Collapse
|
35
|
Mori F, Tanji K, Miki Y, Toyoshima Y, Yoshida M, Kakita A, Takahashi H, Utsumi J, Sasaki H, Wakabayashi K. G protein-coupled receptor 26 immunoreactivity in intranuclear inclusions associated with polyglutamine and intranuclear inclusion body diseases. Neuropathology 2015; 36:50-5. [DOI: 10.1111/neup.12237] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 07/16/2015] [Accepted: 07/16/2015] [Indexed: 01/04/2023]
Affiliation(s)
- Fumiaki Mori
- Department of Neuropathology; Institute of Brain Science, Hirosaki University Graduate School of Medicine; Hirosaki
| | - Kunikazu Tanji
- Department of Neuropathology; Institute of Brain Science, Hirosaki University Graduate School of Medicine; Hirosaki
| | - Yasuo Miki
- Department of Neuropathology; Institute of Brain Science, Hirosaki University Graduate School of Medicine; Hirosaki
| | - Yasuko Toyoshima
- Department of Pathology; Brain Research Institute, University of Niigata; Niigata
| | - Mari Yoshida
- Department of Neuropathology; Aichi Medical University; Nagakute
| | - Akiyoshi Kakita
- Department of Pathological Neuroscience, Center for Bioresource-based Researches; Brain Research Institute, University of Niigata; Niigata
| | - Hitoshi Takahashi
- Department of Pathology; Brain Research Institute, University of Niigata; Niigata
| | - Jun Utsumi
- Department of Neurology; Hokkaido University Graduate School of Medicine; Sapporo Japan
| | - Hidenao Sasaki
- Department of Neurology; Hokkaido University Graduate School of Medicine; Sapporo Japan
| | - Koichi Wakabayashi
- Department of Neuropathology; Institute of Brain Science, Hirosaki University Graduate School of Medicine; Hirosaki
| |
Collapse
|
36
|
Environmental Enrichment Reduces Anxiety by Differentially Activating Serotonergic and Neuropeptide Y (NPY)-Ergic System in Indian Field Mouse (Mus booduga): An Animal Model of Post-Traumatic Stress Disorder. PLoS One 2015; 10:e0127945. [PMID: 26016844 PMCID: PMC4446351 DOI: 10.1371/journal.pone.0127945] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 04/20/2015] [Indexed: 11/19/2022] Open
Abstract
Exposure to a predator elicits an innate fear response and mimics several behavioral disorders related to post-traumatic stress disorder (PTSD). The protective role of an enriched condition (EC) against psychogenic stressors in various animal models has been well documented. However, this condition has not been tested in field mice in the context of PTSD. In this study, we show that field mice (Mus booduga) housed under EC exhibit predominantly proactive and less reactive behavior compared with mice housed under standard conditions (SC) during exposure to their natural predator (field rat Rattus rattus). Furthermore, we observed that EC mice displayed less anxiety-like behavior in an elevated plus maze (EPM) and light/dark-box after exposure to the predator (7 hrs/7 days). In EC mice, predator exposure elevated the level of serotonin (5-Hydroxytrypamine, [5-HT]) in the amygdala as part of the coping response. Subsequently, the serotonin transporter (SERT) and 5-HT1A receptor were up-regulated significantly, but the same did not occur in the 5-HT2C receptor, which is associated with the activation of calmodulin-dependent protein kinase-II (CaMKII) and a transcription factor cAMP response element binding protein (CREB). Our results show that predator exposure induced the activation of CaMKII/CREB, which is accompanied with increased levels of histone acetylation (H3, H4) and decreased histone deacetylases (HDAC1, 2). Subsequently, in the amygdala, the transcription of brain-derived neurotrophic factor (BDNF), neuropeptide Y (NPY) and its Y1 receptor were up-regulated, whereas the Y2 receptor was down-regulated. Therefore, EC facilitated a coping response against a fear associated cue in a PTSD animal model and reduced anxiety by differentially activating serotonergic and NPY-ergic systems.
Collapse
|
37
|
Xu X, Dong F, Yang Y, Wang Y, Wang R, Shen X. Sex-specific effects of long-term exposure to bisphenol-A on anxiety- and depression-like behaviors in adult mice. CHEMOSPHERE 2015; 120:258-266. [PMID: 25112706 DOI: 10.1016/j.chemosphere.2014.07.021] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 07/01/2014] [Accepted: 07/03/2014] [Indexed: 06/03/2023]
Abstract
Humans are routinely exposed to low levels of bisphenol A (BPA), an environmental endocrine disruptor, which is widely used in the production of polycarbonate plastics. The effects of perinatal exposure to BPA have been shown to affect various aspects of social behaviors such as anxiety and depression in adult offspring. Because sex hormones play a critical role in neurobehavior in adulthood, it is possible that long-term exposure to BPA has widespread effects on these emotional behaviors in adulthood. In the present study, adult mice were exposed to BPA at dosages of 0.04, 0.4, 4, 40 mg kg(-1)d(-1) for 12 weeks. A behavioral assay was performed using the open field test (OFT), mirrored maze, the elevated plus maze (EPM), and the forced swim task. The results showed that, after exposure to BPA at 0.4-40 mg kg(-1)d(-1), the number of open arm entries and the time spent in them in the elevated plus maze task were reduced in males but increased in females, and thus eliminating or reversing sex differences in these behaviors. BPA at 0.04-40 mg kg(-1)d(-1) increased the immobility of male mice in the forced swimming test. Furthermore, BPA (0.4-40 mg kg(-1)d(-1)) significantly decreased brain level of testosterone in males, but no significant influence was found in serum and the brain levels of estradiol in females. Western blot analysis further indicated that BPA at 0.4, 4, or 40 mg kg(-1)d(-1) significantly down-regulated the protein level of estrogen receptor β (ERβ) in the hippocampus of the adult males but not females, and inhibited the protein level of GABA(A)α2 receptor in hippocampus of males but promoted that of females. These results suggest that long-term exposure to BPA sex specifically affects anxiety- and depression-like behaviors in adult mice. Changes in the levels of GABA(A)α2 receptor and ERβ proteins of hippocampus might be associated with BPA-induced changes in these emotional behaviors.
Collapse
Affiliation(s)
- Xiaohong Xu
- Institute of Ecology, Chemistry and Life Sciences College, Zhejiang Normal University, China.
| | - Fangni Dong
- Institute of Ecology, Chemistry and Life Sciences College, Zhejiang Normal University, China
| | - Yanling Yang
- Institute of Ecology, Chemistry and Life Sciences College, Zhejiang Normal University, China
| | - Yu Wang
- Institute of Ecology, Chemistry and Life Sciences College, Zhejiang Normal University, China
| | - Ran Wang
- Institute of Ecology, Chemistry and Life Sciences College, Zhejiang Normal University, China
| | - Xiuying Shen
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, China
| |
Collapse
|
38
|
Ahmad R, Wojciech S, Jockers R. Hunting for the function of orphan GPCRs - beyond the search for the endogenous ligand. Br J Pharmacol 2014; 172:3212-28. [PMID: 25231237 DOI: 10.1111/bph.12942] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 09/04/2014] [Accepted: 09/09/2014] [Indexed: 12/13/2022] Open
Abstract
Seven transmembrane-spanning proteins (7TM), also called GPCRs, are among the most versatile and evolutionary successful protein families. Out of the 400 non-odourant members identified in the human genome, approximately 100 remain orphans that have not been matched with an endogenous ligand. Apart from the classical deorphanization strategies, several alternative strategies provided recent new insights into the function of these proteins, which hold promise for high therapeutic potential. These alternative strategies consist of the phenotypical characterization of organisms silenced or overexpressing orphan 7TM proteins, the search for constitutive receptor activity and formation of protein complexes including 7TM proteins as well as the development of synthetic, surrogate ligands. Taken together, a variety of ligand-independent functions can be attributed to orphan 7TM proteins that range from constitutive activity to complex formation with other proteins and include 'true' orphans for which no ligand exist and 'conditional' orphans that behave like orphans in the absence of ligand and as non-orphans in the presence of ligand.
Collapse
Affiliation(s)
- Raise Ahmad
- Institut Cochin, INSERM, Paris, France.,CNRS UMR 8104, Paris, France.,Paris Descartes University, Paris, France
| | - Stefanie Wojciech
- Institut Cochin, INSERM, Paris, France.,CNRS UMR 8104, Paris, France.,Paris Descartes University, Paris, France
| | - Ralf Jockers
- Institut Cochin, INSERM, Paris, France.,CNRS UMR 8104, Paris, France.,Paris Descartes University, Paris, France
| |
Collapse
|
39
|
Xu Y, Wang C, Klabnik JJ, O'Donnell JM. Novel therapeutic targets in depression and anxiety: antioxidants as a candidate treatment. Curr Neuropharmacol 2014; 12:108-19. [PMID: 24669206 PMCID: PMC3964743 DOI: 10.2174/1570159x11666131120231448] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 08/13/2013] [Accepted: 11/02/2013] [Indexed: 01/08/2023] Open
Abstract
There is growing evidence that the imbalance between oxidative stress and the antioxidant defense system may be associated with the development neuropsychiatric disorders, such as depression and anxiety. Major depression and anxiety are presently correlated with a lowered total antioxidant state and by an activated oxidative stress (OS) pathway. The classical antidepressants may produce therapeutic effects other than regulation of monoamines by increasing the antioxidant levels and normalizing the damage caused by OS processes. This chapter provides an overview of recent work on oxidative stress markers in the animal models of depression and anxiety, as well as patients with the aforementioned mood disorders. It is well documented that antioxidants can remove the reactive oxygen species (ROS) and reactive nitrogen species (RNS) through scavenging radicals and suppressing the OS pathway, which further protect against neuronal damage caused oxidative or nitrosative stress sources in the brain, hopefully resulting in remission of depression or anxiety symptoms. The functional understanding of the relationship between oxidative stress and depression and anxiety may pave the way for discovery of novel targets for treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Ying Xu
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Chuang Wang
- Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, PR China
| | - Jonathan J Klabnik
- Department of Behavioral Medicine and Psychiatry, West Virginia University, Morgantown, WV 26508, USA
| | - James M O'Donnell
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
| |
Collapse
|
40
|
Zamora-Martinez ER, Edwards S. Neuronal extracellular signal-regulated kinase (ERK) activity as marker and mediator of alcohol and opioid dependence. Front Integr Neurosci 2014; 8:24. [PMID: 24653683 PMCID: PMC3949304 DOI: 10.3389/fnint.2014.00024] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 02/19/2014] [Indexed: 11/13/2022] Open
Abstract
Early pioneering work in the field of biochemistry identified phosphorylation as a crucial post-translational modification of proteins with the ability to both indicate and arbitrate complex physiological processes. More recent investigations have functionally linked phosphorylation of extracellular signal-regulated kinase (ERK) to a variety of neurophysiological mechanisms ranging from acute neurotransmitter action to long-term gene expression. ERK phosphorylation serves as an intracellular bridging mechanism that facilitates neuronal communication and plasticity. Drugs of abuse, including alcohol and opioids, act as artificial yet powerful rewards that impinge upon natural reinforcement processes critical for survival. The graded progression from initial exposure to addiction (or substance dependence) is believed to result from drug- and drug context-induced adaptations in neuronal signaling processes across brain reward and stress circuits following excessive drug use. In this regard, commonly abused drugs as well as drug-associated experiences are capable of modifying the phosphorylation of ERK within central reinforcement systems. In addition, chronic drug and alcohol exposure may drive ERK-regulated epigenetic and structural alterations that underlie a long-term propensity for escalating drug use. Under the influence of such a neurobiological vulnerability, encountering drug-associated cues and contexts can produce subsequent alterations in ERK signaling that drive relapse to drug and alcohol seeking. Current studies are determining precisely which molecular and regional ERK phosphorylation-associated events contribute to the addiction process, as well as which neuroadaptations need to be targeted in order to return dependent individuals to a healthy state.
Collapse
Affiliation(s)
- Eva R Zamora-Martinez
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute La Jolla, CA, USA
| | - Scott Edwards
- Department of Physiology and Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center New Orleans, LA, USA
| |
Collapse
|
41
|
Hung CJ, Wu CC, Chen WY, Chang CY, Kuan YH, Pan HC, Liao SL, Chen CJ. Depression-like effect of prenatal buprenorphine exposure in rats. PLoS One 2013; 8:e82262. [PMID: 24367510 PMCID: PMC3867331 DOI: 10.1371/journal.pone.0082262] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 11/01/2013] [Indexed: 12/20/2022] Open
Abstract
Studies indicate that perinatal opioid exposure produces a variety of short- and long-term neurobehavioral consequences. However, the precise modes of action are incompletely understood. Buprenorphine, a mixed agonist/antagonist at the opioid receptors, is currently being used in clinical trials for managing pregnant opioid addicts. This study provides evidence of depression-like consequence following prenatal exposure to supra-therapeutic dose of buprenorphine and sheds light on potential mechanisms of action in a rat model involving administration of intraperitoneal injection to pregnant Sprague-Dawley rats starting from gestation day 7 and lasting for 14 days. Results showed that pups at postnatal day 21 but not the dams had worse parameters of depression-like neurobehaviors using a forced swimming test and tail suspension test, independent of gender. Neurobehavioral changes were accompanied by elevation of oxidative stress, reduction of plasma levels of brain-derived neurotrophic factor (BDNF) and serotonin, and attenuation of tropomyosin-related kinase receptor type B (TrkB) phosphorylation, extracellular signal-regulated kinase (ERK) phosphorylation, protein kinase A activity, cAMP response element-binding protein (CREB) phosphorylation, and CREB DNA-binding activity. Since BDNF/serotonin and CREB signaling could orchestrate a positive feedback loop, our findings suggest that the induction of oxidative stress, reduction of BDNF and serotonin expression, and attenuation of CREB signaling induced by prenatal exposure to supra-therapeutic dose of buprenorphine provide evidence of potential mechanism for the development of depression-like neurobehavior.
Collapse
Affiliation(s)
- Chih-Jen Hung
- Department of Anesthesiology, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
- Graduate School of Nursing, HungKuang University, Taichung, Taiwan, ROC
| | - Chih-Cheng Wu
- Department of Anesthesiology, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
- Department of Financial and Computational Mathematics, Providence University, Taichung, Taiwan, ROC
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan, ROC
| | - Cheng-Yi Chang
- Department of Surgery, Fong Yuan Hospital, Taichung, Taiwan, ROC
| | - Yu-Hsiang Kuan
- Department of Pharmacology, Chung Shan Medical University, Taichung, Taiwan, ROC
| | - Hung-Chuan Pan
- Department of Neurosurgery, Research, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Su-Lan Liao
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
| | - Chun-Jung Chen
- Graduate School of Nursing, HungKuang University, Taichung, Taiwan, ROC
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan, ROC
- Center for General Education, Tunghai University, Taichung, Taiwan, ROC
| |
Collapse
|
42
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: G protein-coupled receptors. Br J Pharmacol 2013; 170:1459-581. [PMID: 24517644 PMCID: PMC3892287 DOI: 10.1111/bph.12445] [Citation(s) in RCA: 509] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. G protein-coupled receptors are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, ion channels, catalytic receptors, nuclear hormone receptors, transporters and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen PH Alexander
- School of Life Sciences, University of Nottingham Medical SchoolNottingham, NG7 2UH, UK
| | - Helen E Benson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Elena Faccenda
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Adam J Pawson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Joanna L Sharman
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | | | - John A Peters
- Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of DundeeDundee, DD1 9SY, UK
| | - Anthony J Harmar
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| |
Collapse
|
43
|
Chen C, Yang JM, Hu TT, Xu TJ, Xu WP, Wei W. Elevated dopamine D2 receptor in prefrontal cortex of CUMS rats is associated with downregulated cAMP-independent signaling pathway. Can J Physiol Pharmacol 2013; 91:750-8. [PMID: 23984873 DOI: 10.1139/cjpp-2012-0399] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Because depression is associated with significant morbidity and functional disability, it is important to reveal the mechanism of action. A variety of studies have suggested the involvement of dopaminergic receptors in the pathophysiological mechanism of non-stress-associated depression-like behavior in rodents. Nevertheless, controversy exists about whether chronic stress acts on dopaminergic receptors in the prefrontal cortex. Thus, we investigated the level of dopamine D2 receptors (DRD2) and the possible mechanisms involved in a chronic unpredictable mild stress (CUMS) rat model of depression. The results showed CUMS-induced, depression-like symptoms in the rat, characterized by reduced sucrose consumption and body mass, and increased duration of immobility in a forced swimming test. Moreover, chronic stress upregulated the expression of DRD2 but downregulated protein kinase A (PKA), transcription factor cAMP response element binding protein (CREB), and phospho-CREB (p-CREB) in the prefrontal cortex, as demonstrated by Western blot. Notably, in the rat model of depression, decreased cyclic adenine monophosphate (cAMP) levels and PKA activity were present at the same time, which is consistent with clinical findings in depressed patients. Our findings suggested that dopaminergic system dysfunction could play a central role in stress-related disorders such as depression.
Collapse
Affiliation(s)
- Cheng Chen
- Institute of Clinical Pharmacology of Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology of Education Ministry, Hefei 230032, Anhui, China
| | | | | | | | | | | |
Collapse
|
44
|
Hsieh YS, Chen PN, Kuo MH, Kuo DY. Neuropeptide Y Y1 receptor knockdown can modify glutathione peroxidase and c-AMP response element-binding protein in phenylpropanolamine-treated rats. Arch Toxicol 2013; 87:469-79. [PMID: 23052195 DOI: 10.1007/s00204-012-0947-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 09/18/2012] [Indexed: 10/27/2022]
Abstract
It has been reported that antioxidative enzymes, neuropeptide Y (NPY), and c-AMP response element-binding protein (CREB) are involved in regulating phenylpropanolamine (PPA)-mediated appetite suppression. Here, we investigated whether Y1 receptor (Y1R) might be involved in this regulation. Rats were daily treated with PPA for 4 days. Changes in the contents of NPY, Y1R, glutathione peroxidase (GP), and CREB were assessed and compared. Results showed that Y1R, GP, and CREB increased, with a maximal increase about 100, 200, and 150 %, respectively, on Day 2. By contrast, NPY decreased with a biggest reduction about 48 % on Day 2 and the pattern of expression during PPA treatment was opposite to those of Y1R, GP, and CREB. Central knockdown (using antisense) or inhibition (using antagonist) of Y1R expression modulated the anorectic response of PPA and the reciprocal regulation between NPY and GP (or CREB), revealing an essential role of Y1R in regulating NPY, GP, and CREB. These results suggest that Y1R participates in the reciprocal regulation of NPY, GP, and CREB in the hypothalamus during PPA treatment in conscious rats. The present results may aid the therapeutic research of PPA and related antiobesity drugs.
Collapse
MESH Headings
- Animals
- Appetite Depressants/pharmacology
- Appetite Regulation/drug effects
- Arginine/analogs & derivatives
- Arginine/pharmacology
- Cyclic AMP Response Element-Binding Protein/metabolism
- Dose-Response Relationship, Drug
- Down-Regulation
- Eating/drug effects
- Gene Knockdown Techniques
- Glutathione Peroxidase/metabolism
- Hypothalamus/drug effects
- Hypothalamus/enzymology
- Injections, Intraventricular
- Male
- Neuropeptide Y/metabolism
- Oligonucleotides, Antisense/administration & dosage
- Phenylpropanolamine/pharmacology
- Rats
- Rats, Wistar
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Neuropeptide/antagonists & inhibitors
- Receptors, Neuropeptide/genetics
- Receptors, Neuropeptide/metabolism
- Signal Transduction/drug effects
- Time Factors
Collapse
Affiliation(s)
- Yih-Shou Hsieh
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University and Chung Shan Medical University Hospital, Taichung City, 40201, Taiwan, ROC
| | | | | | | |
Collapse
|
45
|
Buniello A, Hardisty-Hughes RE, Pass JC, Bober E, Smith RJ, Steel KP. Headbobber: a combined morphogenetic and cochleosaccular mouse model to study 10qter deletions in human deafness. PLoS One 2013; 8:e56274. [PMID: 23457544 PMCID: PMC3572983 DOI: 10.1371/journal.pone.0056274] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 01/08/2013] [Indexed: 02/07/2023] Open
Abstract
The recessive mouse mutant headbobber (hb) displays the characteristic behavioural traits associated with vestibular defects including headbobbing, circling and deafness. This mutation was caused by the insertion of a transgene into distal chromosome 7 affecting expression of native genes. We show that the inner ear of hb/hb mutants lacks semicircular canals and cristae, and the saccule and utricle are fused together in a single utriculosaccular sac. Moreover, we detect severe abnormalities of the cochlear sensory hair cells, the stria vascularis looks severely disorganised, Reissner's membrane is collapsed and no endocochlear potential is detected. Myo7a and Kcnj10 expression analysis show a lack of the melanocyte-like intermediate cells in hb/hb stria vascularis, which can explain the absence of endocochlear potential. We use Trp2 as a marker of melanoblasts migrating from the neural crest at E12.5 and show that they do not interdigitate into the developing strial epithelium, associated with abnormal persistence of the basal lamina in the hb/hb cochlea. We perform array CGH, deep sequencing as well as an extensive expression analysis of candidate genes in the headbobber region of hb/hb and littermate controls, and conclude that the headbobber phenotype is caused by: 1) effect of a 648 kb deletion on distal Chr7, resulting in the loss of three protein coding genes (Gpr26, Cpmx2 and Chst15) with expression in the inner ear but unknown function; and 2) indirect, long range effect of the deletion on the expression of neighboring genes on Chr7, associated with downregulation of Hmx3, Hmx2 and Nkx1.2 homeobox transcription factors. Interestingly, deletions of the orthologous region in humans, affecting the same genes, have been reported in nineteen patients with common features including sensorineural hearing loss and vestibular problems. Therefore, we propose that headbobber is a useful model to gain insight into the mechanisms underlying deafness in human 10qter deletion syndrome.
Collapse
Affiliation(s)
- Annalisa Buniello
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, United Kingdom
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | | | - Johanna C. Pass
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, United Kingdom
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - Eva Bober
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Karen P. Steel
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, United Kingdom
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
- MRC Institute of Hearing Research, Nottingham, United Kingdom
- * E-mail:
| |
Collapse
|
46
|
Chen D, Liu X, Zhang W, Shi Y. Targeted inactivation of GPR26 leads to hyperphagia and adiposity by activating AMPK in the hypothalamus. PLoS One 2012; 7:e40764. [PMID: 22815809 PMCID: PMC3398048 DOI: 10.1371/journal.pone.0040764] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 06/12/2012] [Indexed: 11/22/2022] Open
Abstract
G-protein coupled receptor 26 (GPR26) is a brain-specific orphan GPCR with high expression in the brain region that controls satiety. Depletion of GPR26 has been shown to increase fat storage in C. elegans, whereas GPR26 deficiency in the hypothalamus is associated with high genetic susceptibility to the onset of obesity in mice. However, the metabolic function of GPR26 in mammals remains elusive. Herein, we investigated a role of GPR26 in regulating energy homeostasis by generating mice with targeted deletion of the GPR26 gene. We show that GPR26 deficiency causes hyperphagia and hypometabolism, leading to early onset of diet-induced obesity. Accordingly, GPR26 deficiency also caused metabolic complications commonly associated with obesity, including glucose intolerance, hyperinsulinemia, and dyslipidemia. Moreover, consistent with hyperphagia in GPR26 null mice, GPR26 deficiency significantly increased hypothalamic activity of AMPK, a key signaling event that stimulates appetite. In further support of a regulatory role of GPR26 in satiety, GPR26 knockout mice also demonstrate hypersensitivity to treatment of rimonabant, an endocannabinoid receptor-1 antagonist commonly used to treat obesity by suppressing appetite in humans. Together, these findings identified a key role of GPR26 as a central regulator of energy homeostasis though modulation of hypothalamic AMPK activation.
Collapse
Affiliation(s)
- Daohong Chen
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
| | - Xiaolei Liu
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
- Department of Pharmacology, School of Pharmaceutical Sciences Central South University, Changsha, China
| | - Weiping Zhang
- Department of Pathophysiology, Second Military Medical University Shanghai, China
| | - Yuguang Shi
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
| |
Collapse
|
47
|
Abstract
The molecular mechanisms underlying the effects of electroconvulsive shock (ECS) therapy, a fast-acting and very effective antidepressant therapy, are poorly understood. Changes related to neuroplasticity, including enhanced adult hippocampal neurogenesis and neuronal arborization, are believed to play an important role in mediating the effects of ECS. Here we show a dynamic upregulation of the scaffold protein tamalin, selectively in the hippocampus of animals subjected to ECS. Interestingly, this gene upregulation is functionally significant because tamalin deletion in mice abrogated ECS-induced neurogenesis in the adult mouse hippocampus. Furthermore, loss of tamalin blunts mossy fiber sprouting and dendritic arborization caused by ECS. These data suggest an essential role for tamalin in ECS-induced adult neuroplasticity and provide new insight into the pathways that are involved in mediating ECS effects.
Collapse
|