1
|
Alitalo O, Kohtala S, Rosenholm M, Saarreharju R, González-Hernández G, Sarparanta M, Rozov S, Rantamäki T. Nitrous oxide induces hypothermia and TrkB activation: Maintenance of body temperature abolishes antidepressant-like effects in mice. Neuropharmacology 2024; 261:110172. [PMID: 39362627 DOI: 10.1016/j.neuropharm.2024.110172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/05/2024]
Abstract
Recent studies indicate that nitrous oxide (N2O), a gaseous anesthetic and an NMDA (N-methyl-D-aspartate) receptor antagonist, produces rapid antidepressant effect in patients suffering from treatment-resistant depression. Our recent work implies that hypothermia and reduced energy expenditure are connected with antidepressant-induced activation of TrkB neurotrophin receptors - a key regulator of synaptic plasticity. In this study, we demonstrate that a brief exposure to N2O leads to a drop in body temperature following the treatment, which is linked to decreased locomotor activity; enhanced slow-wave electroencephalographic activity; reduced brain glucose utilization; and increased phosphorylation of TrkB, GSK3β (glycogen synthase kinase 3β), and p70S6K (a kinase downstream of mTor (mammalian target of rapamycin)) in the medial prefrontal cortex of adult male mice. Moreover, preventing the hypothermic response in a chronic corticosterone stress model of depression attenuated the antidepressant-like behavioral effects of N2O in the saccharin preference test. These findings indicate that N2O treatment modulates TrkB signaling and related neurotrophic signaling pathways in a temperature-dependent manner, suggesting that the phenomenon driving TrkB activation - altered thermoregulation and energy expenditure - is linked to antidepressant-like behavioral responses.
Collapse
Affiliation(s)
- Okko Alitalo
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland; SleepWell Research Program, Faculty of Medicine, University of Helsinki, Finland
| | - Samuel Kohtala
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland; SleepWell Research Program, Faculty of Medicine, University of Helsinki, Finland
| | - Marko Rosenholm
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland; SleepWell Research Program, Faculty of Medicine, University of Helsinki, Finland; Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Roosa Saarreharju
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland; SleepWell Research Program, Faculty of Medicine, University of Helsinki, Finland
| | - Gemma González-Hernández
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland; SleepWell Research Program, Faculty of Medicine, University of Helsinki, Finland; Neuropsychopharmacology and Psychobiology Research Group, Department of Neurosciences, University of Cádiz, Cádiz, Spain
| | | | - Stanislav Rozov
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland; SleepWell Research Program, Faculty of Medicine, University of Helsinki, Finland
| | - Tomi Rantamäki
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland; SleepWell Research Program, Faculty of Medicine, University of Helsinki, Finland.
| |
Collapse
|
2
|
Ampuero E, Luarte A, Flores FS, Soto AI, Pino C, Silva V, Erlandsen M, Concha T, Wyneken U. The multifaceted effects of fluoxetine treatment on cognitive functions. Front Pharmacol 2024; 15:1412420. [PMID: 39081952 PMCID: PMC11286485 DOI: 10.3389/fphar.2024.1412420] [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: 04/04/2024] [Accepted: 06/10/2024] [Indexed: 08/02/2024] Open
Abstract
Fluoxetine, the prototypical selective serotonin reuptake inhibitor (SSRI), is widely used to treat major depressive disorder (MDD) and a variety of other central nervous system conditions, primarily due to its established clinical safety profile. Although its efficacy in treating depression is well-recognized, the impact of fluoxetine on cognitive functions remains inconsistent and elusive. In this review, we first examine the well-substantiated biological mechanisms underlying fluoxetine's antidepressant effects, which include serotonin reuptake inhibition and activation of TrkB receptors-key to brain-derived neurotrophic factor (BDNF) signaling. Subsequently, we delve into the cognitive side effects observed in both preclinical and clinical studies, affecting domains such as memory, attention, and executive functions. While certain studies indicate cognitive improvements in patients with underlying disorders, there is also evidence of negative effects, influenced by variables like gender, duration of treatment, age, disease pathology, and the specifics of cognitive testing. Significantly, the negative cognitive outcomes reported in preclinical research often involve healthy, non-diseased animals. This review underscores the necessity for heightened caution in fluoxetine prescription and further investigation into its potentially detrimental cognitive effects, even when used prophylactically.
Collapse
Affiliation(s)
- Estíbaliz Ampuero
- Laboratorio Neurofarmacología del Comportamiento, Facultad de Química y Biología, Universidad de Santiago, Santiago, Chile
| | - Alejandro Luarte
- Laboratorio Neurociencias, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Francisca Sofia Flores
- Laboratorio Neurociencias, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Antonia Ignacia Soto
- Laboratorio Neurociencias, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Catalina Pino
- Laboratorio Neurociencias, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Viviana Silva
- Laboratorio Neurociencias, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Macarena Erlandsen
- Laboratorio Neurociencias, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Teresita Concha
- Laboratorio Neurociencias, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Ursula Wyneken
- Laboratorio Neurociencias, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| |
Collapse
|
3
|
Gold PW, Wong ML. Comment on: Antidepressants act by directly binding to TRKB neurotrophin receptors. Mol Psychiatry 2024:10.1038/s41380-024-02615-4. [PMID: 38909095 DOI: 10.1038/s41380-024-02615-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/29/2024] [Accepted: 05/15/2024] [Indexed: 06/24/2024]
Affiliation(s)
- Philip W Gold
- National Institutes of Health, National Institute of Mental Health Intramural Research Program, Bethesda, MD, 20814, USA.
| | - Ma-Li Wong
- State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
4
|
Riggs LM, Pereira EFR, Thompson SM, Gould TD. cAMP-dependent protein kinase signaling is required for ( 2R,6R)-hydroxynorketamine to potentiate hippocampal glutamatergic transmission. J Neurophysiol 2024; 131:64-74. [PMID: 38050689 PMCID: PMC11286304 DOI: 10.1152/jn.00326.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/10/2023] [Accepted: 11/23/2023] [Indexed: 12/06/2023] Open
Abstract
(2R,6R)-Hydroxynorketamine (HNK) is a ketamine metabolite that shows rapid antidepressant-like effects in preclinical studies and lacks the adverse N-methyl-d-aspartate receptor (NMDAR) inhibition-related properties of ketamine. Investigating how (2R,6R)-HNK exerts its antidepressant actions may be informative in the design of novel pharmacotherapies with improved safety and efficacy. We sought to identify the molecular substrates through which (2R,6R)-HNK induces functional changes at excitatory synapses, a prevailing hypothesis for how rapid antidepressant effects are initiated. We recorded excitatory postsynaptic potentials in hippocampal slices from male Wistar Kyoto rats, which have impaired hippocampal plasticity and are resistant to traditional antidepressants. (2R,6R)-HNK (10 µM) led to a rapid potentiation of electrically evoked excitatory postsynaptic potentials at Schaffer collateral CA1 stratum radiatum synapses. This potentiation was associated with a decrease in paired pulse facilitation, suggesting an increase in the probability of glutamate release. The (2R,6R)-HNK-induced potentiation was blocked by inhibiting either cyclic adenosine monophosphate (cAMP) or its downstream target, cAMP-dependent protein kinase (PKA). As cAMP is a potent regulator of brain-derived neurotrophic factor (BDNF) release, we assessed whether (2R,6R)-HNK exerts this acute potentiation through a rapid increase in cAMP-dependent BDNF-TrkB signaling. We found that the cAMP-PKA-dependent potentiation was not dependent on TrkB activation by BDNF, which functionally delimits the acute synaptic effects of (2R,6R)-HNK from its sustained BDNF-dependent actions in vivo. These results suggest that, by potentiating glutamate release via cAMP-PKA signaling, (2R,6R)-HNK initiates acute adaptations in fast excitatory synaptic transmission that promote structural plasticity leading to maintained antidepressant action.NEW & NOTEWORTHY Ketamine is a rapid-acting antidepressant and its preclinical effects are mimicked by its (2R,6R)-(HNK) metabolite. We found that (2R,6R)-HNK initiates acute adaptations in fast excitatory synaptic transmission by potentiating glutamate release via cAMP-PKA signaling at hippocampal Schaffer collateral synapses. This cAMP-PKA-dependent potentiation was not dependent on TrkB activation by BDNF, which functionally delimits the rapid synaptic effects of (2R,6R)-HNK from its sustained BDNF-dependent actions that are thought to maintain antidepressant action in vivo.
Collapse
Affiliation(s)
- Lace M Riggs
- Program in Neuroscience and Training Program in Integrative Membrane Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Edna F R Pereira
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Scott M Thompson
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Veterans Affairs Maryland Health Care System, Baltimore, Maryland, United States
| |
Collapse
|
5
|
Alitalo O, González-Hernández G, Rosenholm M, Kohtala P, Matsui N, Müller HK, Theilmann W, Klein A, Kärkkäinen O, Rozov S, Rantamäki T, Kohtala S. Linking Hypothermia and Altered Metabolism with TrkB Activation. ACS Chem Neurosci 2023; 14:3212-3225. [PMID: 37551888 PMCID: PMC10485900 DOI: 10.1021/acschemneuro.3c00350] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 07/13/2023] [Indexed: 08/09/2023] Open
Abstract
Many mechanisms have been proposed to explain acute antidepressant drug-induced activation of TrkB neurotrophin receptors, but several questions remain. In a series of pharmacological experiments, we observed that TrkB activation induced by antidepressants and several other drugs correlated with sedation, and most importantly, coinciding hypothermia. Untargeted metabolomics of pharmacologically dissimilar TrkB activating treatments revealed effects on shared bioenergetic targets involved in adenosine triphosphate (ATP) breakdown and synthesis, demonstrating a common perturbation in metabolic activity. Both activation of TrkB signaling and hypothermia were recapitulated by administration of inhibitors of glucose and lipid metabolism, supporting a close relationship between metabolic inhibition and neurotrophic signaling. Drug-induced TrkB phosphorylation was independent of electroencephalography slow-wave activity and remained unaltered in knock-in mice with the brain-derived neurotrophic factor (BDNF) Val66Met allele, which have impaired activity-dependent BDNF release, alluding to an activation mechanism independent from BDNF and neuronal activity. Instead, we demonstrated that the active maintenance of body temperature prevents activation of TrkB and other targets associated with antidepressants, including p70S6 kinase downstream of the mammalian target of rapamycin (mTOR) and glycogen synthase kinase 3β (GSK3β). Increased TrkB, GSK3β, and p70S6K phosphorylation was also observed during recovery sleep following sleep deprivation, when a physiological temperature drop is known to occur. Our results suggest that the changes in bioenergetics and thermoregulation are causally connected to TrkB activation and may act as physiological regulators of signaling processes involved in neuronal plasticity.
Collapse
Affiliation(s)
- Okko Alitalo
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
| | - Gemma González-Hernández
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
| | - Marko Rosenholm
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
- Center
for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Piia Kohtala
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
- Department
of Psychiatry, Weill Cornell Medicine, New York, New York 10021, United States
| | - Nobuaki Matsui
- Faculty
of Pharmacy, Gifu University of Medical
Science, 4-3-3 Nijigaoka,
Kani, Gifu 509-0293, Japan
| | - Heidi Kaastrup Müller
- Translational
Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus N 8200, Denmark
| | - Wiebke Theilmann
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - Anders Klein
- Novo
Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen DK-2200, Denmark
- Department
of Drug Design & Pharmacology, University
of Copenhagen, Copenhagen DK-2100, Denmark
| | - Olli Kärkkäinen
- School
of Pharmacy, University of Eastern Finland, Kuopio 70210, Finland
- Afekta
Technologies Ltd., Kuopio 70210, Finland
| | - Stanislav Rozov
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
| | - Tomi Rantamäki
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
| | - Samuel Kohtala
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
- Department
of Psychiatry, Weill Cornell Medicine, New York, New York 10021, United States
| |
Collapse
|
6
|
Sharma V, Singh TG, Kaur A, Mannan A, Dhiman S. Brain-Derived Neurotrophic Factor: A Novel Dynamically Regulated Therapeutic Modulator in Neurological Disorders. Neurochem Res 2023; 48:317-339. [PMID: 36308619 DOI: 10.1007/s11064-022-03755-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 02/04/2023]
Abstract
The growth factor brain-derived neurotrophic factor (BDNF), and its receptor tropomyosin-related kinase receptor type B (TrkB) play an active role in numerous areas of the adult brain, where they regulate the neuronal activity, function, and survival. Upregulation and downregulation of BDNF expression are critical for the physiology of neuronal circuits and functioning in the brain. Loss of BDNF function has been reported in the brains of patients with neurodegenerative or psychiatric disorders. This article reviews the BDNF gene structure, transport, secretion, expression and functions in the brain. This article also implicates BDNF in several brain-related disorders, including Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, major depressive disorder, schizophrenia, epilepsy and bipolar disorder.
Collapse
Affiliation(s)
- Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India.
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India
| | - Sonia Dhiman
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India
| |
Collapse
|
7
|
Jakaria M, Belaidi AA, Southon A, Dent KA, Lane DJR, Bush AI, Ayton S. Receptor-Independent Anti-Ferroptotic Activity of TrkB Modulators. Int J Mol Sci 2022; 23:ijms232416205. [PMID: 36555849 PMCID: PMC9784883 DOI: 10.3390/ijms232416205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/05/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Dysregulated brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB) signalling is implicated in several neurodegenerative diseases, including Alzheimer's disease. A failure of neurotrophic support may participate in neurodegenerative mechanisms, such as ferroptosis, which has likewise been implicated in this disease class. The current study investigated whether modulators of TrkB signalling affect ferroptosis. Cell viability, C11 BODIPY, and cell-free oxidation assays were used to observe the impact of TrkB modulators, and an immunoblot assay was used to detect TrkB expression. TrkB modulators such as agonist BDNF, antagonist ANA-12, and inhibitor K252a did not affect RSL3-induced ferroptosis sensitivity in primary cortical neurons expressing detectable TrkB receptors. Several other modulators of the TrkB receptor, including agonist 7,8-DHF, activator phenelzine sulphate, and inhibitor GNF-5837, conferred protection against a range of ferroptosis inducers in several immortalised neuronal and non-neuronal cell lines, such as N27 and HT-1080 cells. We found these immortalised cell lines lack detectable TrkB receptor expression, so the anti-ferroptotic activity of these TrkB modulators was most likely due to their inherent radical-trapping antioxidant properties, which should be considered when interpreting their experimental findings. These modulators or their variants could be potential anti-ferroptotic therapeutics for various diseases.
Collapse
|
8
|
Suseelan S, Pinna G. Heterogeneity in major depressive disorder: The need for biomarker-based personalized treatments. Adv Clin Chem 2022; 112:1-67. [PMID: 36642481 DOI: 10.1016/bs.acc.2022.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Major Depressive Disorder (MDD) or depression is a pathological mental condition affecting millions of people worldwide. Identification of objective biological markers of depression can provide for a better diagnostic and intervention criteria; ultimately aiding to reduce its socioeconomic health burden. This review provides a comprehensive insight into the major biomarker candidates that have been implicated in depression neurobiology. The key biomarker categories are covered across all the "omics" levels. At the epigenomic level, DNA-methylation, non-coding RNA and histone-modifications have been discussed in relation to depression. The proteomics system shows great promise with inflammatory markers as well as growth factors and neurobiological alterations within the endocannabinoid system. Characteristic lipids implicated in depression together with the endocrine system are reviewed under the metabolomics section. The chapter also examines the novel biomarkers for depression that have been proposed by studies in the microbiome. Depression affects individuals differentially and explicit biomarkers identified by robust research criteria may pave the way for better diagnosis, intervention, treatment, and prediction of treatment response.
Collapse
Affiliation(s)
- Shayam Suseelan
- The Psychiatric Institute, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, United States
| | - Graziano Pinna
- The Psychiatric Institute, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, United States; UI Center on Depression and Resilience (UICDR), Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, United States; Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, United States.
| |
Collapse
|
9
|
Cubillos S, Engmann O, Brancato A. BDNF as a Mediator of Antidepressant Response: Recent Advances and Lifestyle Interactions. Int J Mol Sci 2022; 23:ijms232214445. [PMID: 36430921 PMCID: PMC9698349 DOI: 10.3390/ijms232214445] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/08/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022] Open
Abstract
Conventional antidepressants are widely employed in several psychiatric and neurologic disorders, yet the mechanisms underlying their delayed and partial therapeutic effects are only gradually being understood. This narrative review provides an up-to-date overview of the interplay between antidepressant treatment and Brain-Derived Neurotrophic Factor (BDNF) signaling. In addition, the impact of nutritional, environmental and physiological factors on BDNF and the antidepressant response is outlined. This review underlines the necessity to include information on lifestyle choices in testing and developing antidepressant treatments in the future.
Collapse
Affiliation(s)
- Susana Cubillos
- Institute for Biochemistry and Biophysics, Friedrich-Schiller-University Jena, 07745 Jena, Germany
| | - Olivia Engmann
- Institute for Biochemistry and Biophysics, Friedrich-Schiller-University Jena, 07745 Jena, Germany
- Correspondence:
| | - Anna Brancato
- Department of Sciences for Health Promotion and Mother and Child Care “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
10
|
Martin KK, Noble DJ, Parvin S, Jang K, Garraway SM. Pharmacogenetic inhibition of TrkB signaling in adult mice attenuates mechanical hypersensitivity and improves locomotor function after spinal cord injury. Front Cell Neurosci 2022; 16:987236. [PMID: 36226073 PMCID: PMC9548551 DOI: 10.3389/fncel.2022.987236] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/01/2022] [Indexed: 11/24/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) signals through tropomyosin receptor kinase B (TrkB), to exert various types of plasticity. The exact involvement of BDNF and TrkB in neuropathic pain states after spinal cord injury (SCI) remains unresolved. This study utilized transgenic TrkBF616 mice to examine the effect of pharmacogenetic inhibition of TrkB signaling, induced by treatment with 1NM-PP1 (1NMP) in drinking water for 5 days, on formalin-induced inflammatory pain, pain hypersensitivity, and locomotor dysfunction after thoracic spinal contusion. We also examined TrkB, ERK1/2, and pERK1/2 expression in the lumbar spinal cord and trunk skin. The results showed that formalin-induced pain responses were robustly attenuated in 1NMP-treated mice. Weekly assessment of tactile sensitivity with the von Frey test showed that treatment with 1NMP immediately after SCI blocked the development of mechanical hypersensitivity up to 4 weeks post-SCI. Contrastingly, when treatment started 2 weeks after SCI, 1NMP reversibly and partially attenuated hind-paw hypersensitivity. Locomotor scores were significantly improved in the early-treated 1NMP mice compared to late-treated or vehicle-treated SCI mice. 1NMP treatment attenuated SCI-induced increases in TrkB and pERK1/2 levels in the lumbar cord but failed to exert similar effects in the trunk skin. These results suggest that early onset TrkB signaling after SCI contributes to maladaptive plasticity that leads to spinal pain hypersensitivity and impaired locomotor function.
Collapse
Affiliation(s)
| | | | | | | | - Sandra M. Garraway
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
11
|
Physical exercise prevents behavioral alterations in a reserpine-treated zebrafish: A putative depression model. Pharmacol Biochem Behav 2022; 220:173455. [PMID: 36063969 DOI: 10.1016/j.pbb.2022.173455] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/20/2022]
Abstract
Major depressive disorder (MDD) has increasingly reached the world population with an expressive increase in recent years due to the COVID-19 pandemic. Here we used adult zebrafish (Danio rerio) as a model to verify the effects of reserpine on behavior and neurotransmitter levels. We observed an increase in the immobile time and time spent in the bottom zone of the tank in reserpine-exposed animals. The results demonstrated a decrease in distance traveled and velocity. Reserpine exposure did not induce changes in memory and social interaction compared to the control group. We also evaluated the influence of exposure to fluoxetine, a well-known antidepressant, on the behavior of reserpine-exposed animals. We observed a reversal of behavioral alterations caused by reserpine. To verify whether behavioral alterations in the putative depression model induced by reserpine could be prevented, the animals were subjected to physical exercise for 6 weeks. The results showed a protective effect of the physical exercise against the behavioral changes caused by reserpine in zebrafish. In addition, we observed a reduction in dopamine and serotonin levels and an increase in the 3,4-dihydroxyphenylacetic acid (DOPAC) levels in the brain. Physical exercise was able to prevent the changes in dopamine and serotonin levels, reinforcing that the preventive effect promoted by physical exercise is related to the modulation of neurotransmitter levels. Our findings showed that reserpine was effective in the induction of a putative depression model in zebrafish and that physical exercise may be an alternative to prevent the effects induced by reserpine.
Collapse
|
12
|
Orciani C, Hall H, Pentz R, Foret MK, Do Carmo S, Cuello AC. Long-term nucleus basalis cholinergic depletion induces attentional deficits and impacts cortical neurons and BDNF levels without affecting the NGF synthesis. J Neurochem 2022; 163:149-167. [PMID: 35921478 DOI: 10.1111/jnc.15683] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 11/26/2022]
Abstract
Basal forebrain cholinergic neurons (BFCNs) represent the main source of cholinergic innervation to the cortex and hippocampus and degenerate early in Alzheimer's disease (AD) progression. Phenotypic maintenance of BFCNs depends on levels of mature nerve growth factor (mNGF) and mature brain-derived neurotrophic factor (mBDNF), produced by target neurons and retrogradely transported to the cell body. Whether a reciprocal interaction where BFCN inputs impact neurotrophin availability and affect cortical neuronal markers is unknown. To address our hypothesis, we immunolesioned the nucleus basalis (nb), a basal forebrain cholinergic nuclei projecting mainly to the cortex, by bilateral stereotaxic injection of 192-IgG-Saporin (the cytotoxin Saporin binds p75ntr receptors expressed exclusively by BFCNs) in 2.5-month-old Wistar rats. At six months post-lesion, Saporin-injected rats (SAP) showed an impairment in a modified version of the 5-Choice Serial Reaction Time Task (5-choice task). Post-mortem analyses of the brain revealed a reduction of Choline Acetyltransferase-immunoreactive neurons compared to wild-type controls. A diminished number of cortical vesicular acetylcholine transporter-immunoreactive boutons was accompanied by a reduction in BDNF mRNA, mBDNF protein levels, markers of glutamatergic (vGluT1) and GABAergic (GAD65) neurons in the SAP-group compared to the controls. NGF mRNA, NGF precursor and mNGF protein levels were not affected. Additionally, cholinergic markers correlated with the attentional deficit and BDNF levels. Our findings demonstrate that while cholinergic nb loss impairs cognition and reduces cortical neuron markers, it produces differential effects on neurotrophin availability, affecting BDNF but not NGF levels.
Collapse
Affiliation(s)
- Chiara Orciani
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Helene Hall
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Rowan Pentz
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Morgan K Foret
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Sonia Do Carmo
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - A Claudio Cuello
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada.,Department of Pharmacology, Oxford University, US (Visiting Professor)
| |
Collapse
|
13
|
Pro-cognitive effect of acute imipramine administration correlates with direct interaction of BDNF with its receptor, Trkβ. Brain Res 2022; 1789:147948. [PMID: 35597327 DOI: 10.1016/j.brainres.2022.147948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/01/2022] [Accepted: 05/15/2022] [Indexed: 01/18/2023]
Abstract
Given the important role of brain-derived neurotrophic factor (BDNF)-mediated Trkβ signalling in the mechanism of action of antidepressants (ADs), we examined ligand-receptor interactions in the rat cingulate cortex using a proximity ligation assay (PLA) in response to acute and repeated administration of imipramine (IMI), followed by various drug-free periods. Both the acute and chronic administration of IMI increased the BDNF-Trkβ interaction observed 3 h after drug administration. Withdrawal of IMI for 72 h or 7 days did not alter BDNF-Trkβ interaction. A significant reduction in this interaction after chronic IMI administration followed by 21 drug-free days was observed, but it returned to the control value when a new dose of IMI was given after this time. The level of mRNA encoding BDNF or Trkβ did not change in the experimental groups of animals, so one can conclude that alterations in the BDNF-Trkβ interaction depend not on acute vs. repeated treatment with IMI but on the presence of the drug in the body. This effect correlates well with the strong pro-cognitive effect of acute IMI, assessed by the novel object recognition (NOR) test.
Collapse
|
14
|
Hess EM, Riggs LM, Michaelides M, Gould TD. Mechanisms of ketamine and its metabolites as antidepressants. Biochem Pharmacol 2022; 197:114892. [PMID: 34968492 PMCID: PMC8883502 DOI: 10.1016/j.bcp.2021.114892] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 02/06/2023]
Abstract
Treating major depression is a medical need that remains unmet by monoaminergic therapeutic strategies that commonly fail to achieve symptom remission. A breakthrough in the treatment of depression was the discovery that the anesthetic (R,S)-ketamine (ketamine), when administered at sub-anesthetic doses, elicits rapid (sometimes within hours) antidepressant effects in humans that are otherwise resistant to monoaminergic-acting therapies. While this finding was revolutionary and led to the FDA approval of (S)-ketamine (esketamine) for use in adults with treatment-resistant depression and suicidal ideation, the mechanisms underlying how ketamine or esketamine elicit their effects are still under active investigation. An emerging view is that metabolism of ketamine may be a crucial step in its mechanism of action, as several metabolites of ketamine have neuroactive effects of their own and may be leveraged as therapeutics. For example, (2R,6R)-hydroxynorketamine (HNK), is readily observed in humans following ketamine treatment and has shown therapeutic potential in preclinical tests of antidepressant efficacy and synaptic potentiation while being devoid of the negative adverse effects of ketamine, including its dissociative properties and abuse potential. We discuss preclinical and clinical studies pertaining to how ketamine and its metabolites produce antidepressant effects. Specifically, we explore effects on glutamate neurotransmission through N-methyl D-aspartate receptors (NMDARs) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs), synaptic structural changes via brain derived neurotrophic factor (BDNF) signaling, interactions with opioid receptors, and the enhancement of serotonin, norepinephrine, and dopamine signaling. Strategic targeting of these mechanisms may result in novel rapid-acting antidepressants with fewer undesirable side effects compared to ketamine.
Collapse
Affiliation(s)
- Evan M Hess
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Lace M Riggs
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA.,Program in Neuroscience and Training Program in Integrative Membrane Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Michael Michaelides
- Biobehavioral Imaging & Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA.,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Departments of Pharmacology and Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Baltimore Veterans Affairs Medical Center, Veterans Affairs Maryland Health Care System, Baltimore, MD 21201, USA.
| |
Collapse
|
15
|
Mezhlumyan AG, Tallerova AV, Povarnina PY, Tarasiuk AV, Sazonova NM, Gudasheva TA, Seredenin SB. Antidepressant-like Effects of BDNF and NGF Individual Loop Dipeptide Mimetics Depend on the Signal Transmission Patterns Associated with Trk. Pharmaceuticals (Basel) 2022; 15:ph15030284. [PMID: 35337082 PMCID: PMC8950955 DOI: 10.3390/ph15030284] [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: 01/20/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 11/29/2022] Open
Abstract
Neurotrophins are considered as an attractive target for the development of antidepressants with a novel mechanism of action. Previously, the dimeric dipeptide mimetics of individual loops of nerve growth factor, NGF (GK-6, loop 1; GK-2, loop 4) and brain-derived neurotrophic factor, BDNF (GSB-214, loop 1; GTS-201, loop 2; GSB-106, loop 4) were designed and synthesized. All the mimetics of NGF and BDNF in vitro after a 5–180 min incubation in a HT-22 cell culture were able to phosphorylate the tropomyosin-related kinase A (TrkA) or B (TrkB) receptors, respectively, but had different post-receptor signaling patterns. In the present study, we conduct comparative research of the antidepressant-like activity of these mimetics at acute and subchronic administration in the forced swim test in mice. Only the dipeptide GSB-106 that in vitro activates mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK), phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) and phospholipase C-gamma (PLCγ) post-receptor pathways exhibited antidepressant-like activity (0.1 and 1.0 mg/kg, ip) at acute administration. At the same time, the inhibition of any one of these signaling pathways completely prevented the antidepressant-like effects of GSB-106 in the forced swim test. All the NGF mimetics were inactive after a single injection regardless of post-receptor in vitro signaling patterns. All the investigated dipeptides, except GTS-201, not activating PI3K/AKT in vitro unlike the other compounds, were active at subchronic administration. The data obtained demonstrate that the low-molecular weight BDNF mimetic GSB-106 that activates all three main post-receptor TrkB signaling pathways is the most promising for the development as an antidepressant.
Collapse
Affiliation(s)
- Armen G. Mezhlumyan
- Department of Medicinal Chemistry, V.V. Zakusov Research Institute of Pharmacology, 125315 Moscow, Russia; (A.G.M.); (A.V.T.); (P.Y.P.); (A.V.T.); (N.M.S.)
| | - Anna V. Tallerova
- Department of Medicinal Chemistry, V.V. Zakusov Research Institute of Pharmacology, 125315 Moscow, Russia; (A.G.M.); (A.V.T.); (P.Y.P.); (A.V.T.); (N.M.S.)
| | - Polina Y. Povarnina
- Department of Medicinal Chemistry, V.V. Zakusov Research Institute of Pharmacology, 125315 Moscow, Russia; (A.G.M.); (A.V.T.); (P.Y.P.); (A.V.T.); (N.M.S.)
| | - Aleksey V. Tarasiuk
- Department of Medicinal Chemistry, V.V. Zakusov Research Institute of Pharmacology, 125315 Moscow, Russia; (A.G.M.); (A.V.T.); (P.Y.P.); (A.V.T.); (N.M.S.)
| | - Nellya M. Sazonova
- Department of Medicinal Chemistry, V.V. Zakusov Research Institute of Pharmacology, 125315 Moscow, Russia; (A.G.M.); (A.V.T.); (P.Y.P.); (A.V.T.); (N.M.S.)
| | - Tatiana A. Gudasheva
- Department of Medicinal Chemistry, V.V. Zakusov Research Institute of Pharmacology, 125315 Moscow, Russia; (A.G.M.); (A.V.T.); (P.Y.P.); (A.V.T.); (N.M.S.)
- Correspondence:
| | - Sergey B. Seredenin
- Department of Pharmacogenetics, V.V. Zakusov Research Institute of Pharmacology, 25315 Moscow, Russia;
| |
Collapse
|
16
|
Analysis of Antidepressant-like Effects and Action Mechanisms of GSB-106, a Small Molecule, Affecting the TrkB Signaling. Int J Mol Sci 2021; 22:ijms222413381. [PMID: 34948177 PMCID: PMC8704497 DOI: 10.3390/ijms222413381] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/01/2021] [Accepted: 12/10/2021] [Indexed: 12/13/2022] Open
Abstract
Induction of BDNF-TrkB signaling is associated with the action mechanisms of conventional and fast-acting antidepressants. GSB-106, developed as a small dimeric dipeptide mimetic of BDNF, was previously shown to produce antidepressant-like effects in the mouse Porsolt test, tail suspension test, Nomura water wheel test, in the chronic social defeat stress model and in the inflammation-induced model of depression. In the present study, we evaluated the effect of chronic per os administration of GSB-106 to Balb/c mice under unpredictable chronic mild stress (UCMS). It was observed for the first time that long term GSB-106 treatment (1 mg/kg, 26 days) during ongoing UCMS procedure ameliorated the depressive-like behaviors in mice as indicated by the Porsolt test. In addition, chronic per os administration of GSB-106 resulted in an increase in BDNF levels, which were found to be decreased in the prefrontal cortex and hippocampus of mice after UCMS. Furthermore, prolonged GSB-106 treatment was accompanied by an increase in the content of pTrkB706/707 in the prefrontal cortex and by a pronounced increase in the level of pTrkB816 in both studied brain structures of mice subjected to UCMS procedure. In summary, the present data show that chronic GSB-106 treatment produces an antidepressant-like effect in the unpredictable chronic mild stress model, which is likely to be associated with the regulation of the BDNF-TrkB signaling.
Collapse
|
17
|
Quinn JP, Kandigian SE, Trombetta BA, Arnold SE, Carlyle BC. VGF as a biomarker and therapeutic target in neurodegenerative and psychiatric diseases. Brain Commun 2021; 3:fcab261. [PMID: 34778762 PMCID: PMC8578498 DOI: 10.1093/braincomms/fcab261] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/01/2021] [Accepted: 09/13/2021] [Indexed: 12/20/2022] Open
Abstract
Neurosecretory protein VGF (non-acronymic) belongs to the granin family of neuropeptides. VGF and VGF-derived peptides have been repeatedly identified in well-powered and well-designed multi-omic studies as dysregulated in neurodegenerative and psychiatric diseases. New therapeutics is urgently needed for these devastating and costly diseases, as are new biomarkers to improve disease diagnosis and mechanistic understanding. From a list of 537 genes involved in Alzheimer's disease pathogenesis, VGF was highlighted by the Accelerating Medicines Partnership in Alzheimer's disease as the potential therapeutic target of greatest interest. VGF levels are consistently decreased in brain tissue and CSF samples from patients with Alzheimer's disease compared to controls, and its levels correlate with disease severity and Alzheimer's disease pathology. In the brain, VGF exists as multiple functional VGF-derived peptides. Full-length human VGF1-615 undergoes proteolytic processing by prohormone convertases and other proteases in the regulated secretory pathway to produce at least 12 active VGF-derived peptides. In cell and animal models, these VGF-derived peptides have been linked to energy balance regulation, neurogenesis, synaptogenesis, learning and memory, and depression-related behaviours throughout development and adulthood. The C-terminal VGF-derived peptides, TLQP-62 (VGF554-615) and TLQP-21 (VGF554-574) have differential effects on Alzheimer's disease pathogenesis, neuronal and microglial activity, and learning and memory. TLQP-62 activates neuronal cell-surface receptors and regulates long-term hippocampal memory formation. TLQP-62 also prevents immune-mediated memory impairment, depression-like and anxiety-like behaviours in mice. TLQP-21 binds to microglial cell-surface receptors, triggering microglial chemotaxis and phagocytosis. These actions were reported to reduce amyloid-β plaques and decrease neuritic dystrophy in a transgenic mouse model of familial Alzheimer's disease. Expression differences of VGF-derived peptides have also been associated with frontotemporal lobar dementias, amyotrophic lateral sclerosis, Lewy body diseases, Huntington's disease, pain, schizophrenia, bipolar disorder, depression and antidepressant response. This review summarizes current knowledge and highlights questions for future investigation regarding the roles of VGF and its dysregulation in neurodegenerative and psychiatric disease. Finally, the potential of VGF and VGF-derived peptides as biomarkers and novel therapeutic targets for neurodegenerative and psychiatric diseases is highlighted.
Collapse
Affiliation(s)
- James P Quinn
- Department of Neurology, Alzheimer's Clinical & Translational Research Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Savannah E Kandigian
- Department of Neurology, Alzheimer's Clinical & Translational Research Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Bianca A Trombetta
- Department of Neurology, Alzheimer's Clinical & Translational Research Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Steven E Arnold
- Department of Neurology, Alzheimer's Clinical & Translational Research Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Becky C Carlyle
- Department of Neurology, Alzheimer's Clinical & Translational Research Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
18
|
Prowse N, Hayley S. Microglia and BDNF at the crossroads of stressor related disorders: Towards a unique trophic phenotype. Neurosci Biobehav Rev 2021; 131:135-163. [PMID: 34537262 DOI: 10.1016/j.neubiorev.2021.09.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/16/2022]
Abstract
Stressors ranging from psychogenic/social to neurogenic/injury to systemic/microbial can impact microglial inflammatory processes, but less is known regarding their effects on trophic properties of microglia. Recent studies do suggest that microglia can modulate neuronal plasticity, possibly through brain derived neurotrophic factor (BDNF). This is particularly important given the link between BDNF and neuropsychiatric and neurodegenerative pathology. We posit that certain activated states of microglia play a role in maintaining the delicate balance of BDNF release onto neuronal synapses. This focused review will address how different "activators" influence the expression and release of microglial BDNF and address the question of tropomyosin receptor kinase B (TrkB) expression on microglia. We will then assess sex-based differences in microglial function and BDNF expression, and how microglia are involved in the stress response and related disorders such as depression. Drawing on research from a variety of other disorders, we will highlight challenges and opportunities for modulators that can shift microglia to a "trophic" phenotype with a view to potential therapeutics relevant for stressor-related disorders.
Collapse
Affiliation(s)
- Natalie Prowse
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada.
| | - Shawn Hayley
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada.
| |
Collapse
|
19
|
Facilitation of TRKB Activation by the Angiotensin II Receptor Type-2 (AT2R) Agonist C21. Pharmaceuticals (Basel) 2021; 14:ph14080773. [PMID: 34451870 PMCID: PMC8400827 DOI: 10.3390/ph14080773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 11/25/2022] Open
Abstract
Blockers of angiotensin II type 1 receptor (AT1R) exert antidepressant-like effects by indirectly facilitating the activation of the angiotensin II type 2 receptor (AT2R), which leads to increased surface expression and transactivation of tropomyosin-related kinase B receptors (TRKB). Compound 21 (C21) is a non-peptide AT2R agonist that produces neuroprotective effects. However, the behavioral effects of C21 and its involvement with the brain-derived neurotrophic factor (BDNF)-TRKB system still need further investigation. The aim of the present study was to assess the effect of C21 on the activation of TRKB and its consequences on conditioned fear. The administration of C21 (0.1–10 μM/15 min) increased the surface levels of TRKB but was not sufficient to increase the levels of phosphorylated TRKB (pTRKB) in cultured cortical neurons from rat embryos. Consistent with increased TRKB surface expression, C21 (10 μM/15 min or 3 days) facilitated the effect of BDNF (0.1 ng/mL/15 min) on pTRKB in these cells. In contextual fear conditioning, the freezing time of C21-treated (administered intranasally) wild-type mice was decreased compared to the vehicle-treated group, but no effect of C21 was observed in BDNF.het animals. We observed no effect of C21 in the elevated plus-maze test for anxiety. Taken together, our results indicate that C21 facilitated BDNF effect by increasing the levels of TRKB on the cell surface and reduced the freezing time of mice in a BDNF-dependent manner, but not through a general anxiolytic-like effect.
Collapse
|
20
|
Serotonin Heteroreceptor Complexes and Their Integration of Signals in Neurons and Astroglia-Relevance for Mental Diseases. Cells 2021; 10:cells10081902. [PMID: 34440670 PMCID: PMC8392445 DOI: 10.3390/cells10081902] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/16/2021] [Accepted: 07/23/2021] [Indexed: 12/28/2022] Open
Abstract
The heteroreceptor complexes present a novel biological principle for signal integration. These complexes and their allosteric receptor-receptor interactions are bidirectional and novel targets for treatment of CNS diseases including mental diseases. The existence of D2R-5-HT2AR heterocomplexes can help explain the anti-schizophrenic effects of atypical antipsychotic drugs not only based on blockade of 5-HT2AR and of D2R in higher doses but also based on blocking the allosteric enhancement of D2R protomer signaling by 5-HT2AR protomer activation. This research opens a new understanding of the integration of DA and 5-HT signals released from DA and 5-HT nerve terminal networks. The biological principle of forming 5-HT and other heteroreceptor complexes in the brain also help understand the mechanism of action for especially the 5-HT hallucinogens, including putative positive effects of e.g., psilocybin and the indicated prosocial and anti-stress actions of MDMA (ecstasy). The GalR1-GalR2 heterodimer and the putative GalR1-GalR2-5-HT1 heteroreceptor complexes are targets for Galanin N-terminal fragment Gal (1-15), a major modulator of emotional networks in models of mental disease. GPCR-receptor tyrosine kinase (RTK) heteroreceptor complexes can operate through transactivation of FGFR1 via allosteric mechanisms and indirect interactions over GPCR intracellular pathways involving protein kinase Src which produces tyrosine phosphorylation of the RTK. The exciting discovery was made that several antidepressant drugs such as TCAs and SSRIs as well as the fast-acting antidepressant drug ketamine can directly bind to the TrkB receptor and provide a novel mechanism for their antidepressant actions. Understanding the role of astrocytes and their allosteric receptor-receptor interactions in modulating forebrain glutamate synapses with impact on dorsal raphe-forebrain serotonin neurons is also of high relevance for research on major depressive disorder.
Collapse
|
21
|
Colombo E, Triolo D, Bassani C, Bedogni F, Di Dario M, Dina G, Fredrickx E, Fermo I, Martinelli V, Newcombe J, Taveggia C, Quattrini A, Comi G, Farina C. Dysregulated copper transport in multiple sclerosis may cause demyelination via astrocytes. Proc Natl Acad Sci U S A 2021; 118:e2025804118. [PMID: 34183414 PMCID: PMC8271600 DOI: 10.1073/pnas.2025804118] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Demyelination is a key pathogenic feature of multiple sclerosis (MS). Here, we evaluated the astrocyte contribution to myelin loss and focused on the neurotrophin receptor TrkB, whose up-regulation on the astrocyte finely demarcated chronic demyelinated areas in MS and was paralleled by neurotrophin loss. Mice lacking astrocyte TrkB were resistant to demyelination induced by autoimmune or toxic insults, demonstrating that TrkB signaling in astrocytes fostered oligodendrocyte damage. In vitro and ex vivo approaches highlighted that astrocyte TrkB supported scar formation and glia proliferation even in the absence of neurotrophin binding, indicating TrkB transactivation in response to inflammatory or toxic mediators. Notably, our neuropathological studies demonstrated copper dysregulation in MS and model lesions and TrkB-dependent expression of copper transporter (CTR1) on glia cells during neuroinflammation. In vitro experiments evidenced that TrkB was critical for the generation of glial intracellular calcium flux and CTR1 up-regulation induced by stimuli distinct from neurotrophins. These events led to copper uptake and release by the astrocyte, and in turn resulted in oligodendrocyte loss. Collectively, these data demonstrate a pathogenic demyelination mechanism via the astrocyte release of copper and open up the possibility of restoring copper homeostasis in the white matter as a therapeutic target in MS.
Collapse
Affiliation(s)
- Emanuela Colombo
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Daniela Triolo
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Claudia Bassani
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Francesco Bedogni
- San Raffaele Rett Research Centre, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Marco Di Dario
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Giorgia Dina
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Evelien Fredrickx
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Isabella Fermo
- Division of Immunology, Transplantation, and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Vittorio Martinelli
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Jia Newcombe
- NeuroResource, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, WC1N 1PJ, London, UK
| | - Carla Taveggia
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Angelo Quattrini
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Giancarlo Comi
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Cinthia Farina
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy;
| |
Collapse
|
22
|
Campos-Cardoso R, Silva CPB, Carolino ROG, Anselmo-Franci JA, Tirapelli CR, Padovan CM. Imipramine attenuates anxiety- and depressive-like effects of acute and prolonged ethanol-abstinence in male rats by modulating SERT and GR expression in the dorsal hippocampus. Behav Brain Res 2021; 408:113295. [PMID: 33839161 DOI: 10.1016/j.bbr.2021.113295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 04/02/2021] [Accepted: 04/05/2021] [Indexed: 10/21/2022]
Abstract
AIMS Considering that serotoninergic agents attenuate symptoms of anxiety and are used to treat depression, we investigated whether subchronic treatment with imipramine, a serotonin/noradrenaline reuptake inhibitor, would prevent the anxiogenic-like behaviour induced by acute and/or chronic ethanol withdrawal. We also investigated whether those changes were related to the disfunctioning of hypothalamic-pituitary-adrenal (HPA) axis and serotonergic neurotransmission. MAIN METHODS 264 Male Wistar rats were treated with ethanol 6% (vol./vol.) for 21 days. Acute ethanol withdrawal was induced by abrupt discontinuation of treatment and sustained for 48 h. Protracted abstinence was sustained for an additional period of 21 days. Behavioural tests included the Elevated Plus Maze (EPM) or Light/Dark Box (LDB) after acute abstinence, and the Forced Swim Test (FST) after protracted abstinence. Imipramine (15 mg/kg, i.p.) was administered 24, 19 and 1 h before EPM or LDB tests. KEY FINDINGS Acute abstinence decreased exploration of the open arms of the EPM, without changing exploration of LDB. Additionally, chronic abstinent rats displayed more time immobile in the FST, when compared to control animals. These effects were attenuated by imipramine treatment, without changing basal response. Imipramine prevented protracted abstinence -induced decrease in glucocorticoid receptor (GR) and serotonin transporter (SERT) expression in the dorsal hippocampus. SIGNIFICANCE Our findings indicate that chronic ethanol withdrawal affects the hippocampal serotonergic system by decreasing serotonin transporter expression. It also disturbs the HPA axis functioning through an imbalance on GR and mineralocorticoid (MR) expression.
Collapse
Affiliation(s)
- Rodrigo Campos-Cardoso
- Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil; Faculdade de Medicina de Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil
| | - Carla P B Silva
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil; Escola de Enfermagem de Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil
| | | | | | - Carlos R Tirapelli
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil; Escola de Enfermagem de Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil; Instituto de Neurociências e Comportamento (INeC), Ribeirão Preto, SP, Brazil
| | - Cláudia M Padovan
- Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil; Faculdade de Medicina de Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil; Instituto de Neurociências e Comportamento (INeC), Ribeirão Preto, SP, Brazil.
| |
Collapse
|
23
|
Kohtala S. Ketamine-50 years in use: from anesthesia to rapid antidepressant effects and neurobiological mechanisms. Pharmacol Rep 2021; 73:323-345. [PMID: 33609274 PMCID: PMC7994242 DOI: 10.1007/s43440-021-00232-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 02/03/2021] [Accepted: 02/06/2021] [Indexed: 12/11/2022]
Abstract
Over the past 50 years, ketamine has solidified its position in both human and veterinary medicine as an important anesthetic with many uses. More recently, ketamine has been studied and used for several new indications, ranging from chronic pain to drug addiction and post-traumatic stress disorder. The discovery of the rapid-acting antidepressant effects of ketamine has resulted in a surge of interest towards understanding the precise mechanisms driving its effects. Indeed, ketamine may have had the largest impact for advancements in the research and treatment of psychiatric disorders in the past few decades. While intense research efforts have been aimed towards uncovering the molecular targets underlying ketamine's effects in treating depression, the underlying neurobiological mechanisms remain elusive. These efforts are made more difficult by ketamine's complex dose-dependent effects on molecular mechanisms, multiple pharmacologically active metabolites, and a mechanism of action associated with the facilitation of synaptic plasticity. This review aims to provide a brief overview of the different uses of ketamine, with an emphasis on examining ketamine's rapid antidepressant effects spanning molecular, cellular, and network levels. Another focus of the review is to offer a perspective on studies related to the different doses of ketamine used in antidepressant research. Finally, the review discusses some of the latest hypotheses concerning ketamine's action.
Collapse
Affiliation(s)
- Samuel Kohtala
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P. O. Box 56, 00014, Helsinki, Finland.
- SleepWell Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Feil Family Brain and Mind Research Institute, Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
24
|
Casarotto PC, Girych M, Fred SM, Kovaleva V, Moliner R, Enkavi G, Biojone C, Cannarozzo C, Sahu MP, Kaurinkoski K, Brunello CA, Steinzeig A, Winkel F, Patil S, Vestring S, Serchov T, Diniz CRAF, Laukkanen L, Cardon I, Antila H, Rog T, Piepponen TP, Bramham CR, Normann C, Lauri SE, Saarma M, Vattulainen I, Castrén E. Antidepressant drugs act by directly binding to TRKB neurotrophin receptors. Cell 2021; 184:1299-1313.e19. [PMID: 33606976 PMCID: PMC7938888 DOI: 10.1016/j.cell.2021.01.034] [Citation(s) in RCA: 352] [Impact Index Per Article: 117.3] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 12/22/2020] [Accepted: 01/21/2021] [Indexed: 12/11/2022]
Abstract
It is unclear how binding of antidepressant drugs to their targets gives rise to the clinical antidepressant effect. We discovered that the transmembrane domain of tyrosine kinase receptor 2 (TRKB), the brain-derived neurotrophic factor (BDNF) receptor that promotes neuronal plasticity and antidepressant responses, has a cholesterol-sensing function that mediates synaptic effects of cholesterol. We then found that both typical and fast-acting antidepressants directly bind to TRKB, thereby facilitating synaptic localization of TRKB and its activation by BDNF. Extensive computational approaches including atomistic molecular dynamics simulations revealed a binding site at the transmembrane region of TRKB dimers. Mutation of the TRKB antidepressant-binding motif impaired cellular, behavioral, and plasticity-promoting responses to antidepressants in vitro and in vivo. We suggest that binding to TRKB and allosteric facilitation of BDNF signaling is the common mechanism for antidepressant action, which may explain why typical antidepressants act slowly and how molecular effects of antidepressants are translated into clinical mood recovery.
Collapse
Affiliation(s)
| | - Mykhailo Girych
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Senem M Fred
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland
| | - Vera Kovaleva
- Institute of Biotechnology-HILIFE, University of Helsinki, Helsinki, Finland
| | - Rafael Moliner
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland
| | - Giray Enkavi
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Caroline Biojone
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland
| | | | | | - Katja Kaurinkoski
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland
| | | | - Anna Steinzeig
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland
| | - Frederike Winkel
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland
| | - Sudarshan Patil
- Department of Biomedicine and KG Jebsen Center for Research on Neuropsychiatric Disorders, University of Bergen, Bergen, Norway
| | - Stefan Vestring
- Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Berta-Ottenstein-Programme for Clinician Scientists, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tsvetan Serchov
- Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Cassiano R A F Diniz
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland; Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paul, Brazil
| | - Liina Laukkanen
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland
| | - Iseline Cardon
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland; Brain Master Program, Faculty of Science, Aix-Marseille Université, Marseille, France; Department of Psychiatry, University of Regensburg, Regenburg, Germany
| | - Hanna Antila
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland; Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tomasz Rog
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Timo Petteri Piepponen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Clive R Bramham
- Department of Biomedicine and KG Jebsen Center for Research on Neuropsychiatric Disorders, University of Bergen, Bergen, Norway
| | - Claus Normann
- Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center for Basics in Neuromodulation (NeuroModul Basics), University of Freiburg, Freiburg, Germany
| | - Sari E Lauri
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland; Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology-HILIFE, University of Helsinki, Helsinki, Finland
| | - Ilpo Vattulainen
- Department of Physics, University of Helsinki, Helsinki, Finland; Computational Physics Laboratory, Tampere University, Tampere, Finland
| | - Eero Castrén
- Neuroscience Center-HILIFE, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
25
|
The Role of Neurotrophic Factors in Pathophysiology of Major Depressive Disorder. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1305:257-272. [PMID: 33834404 DOI: 10.1007/978-981-33-6044-0_14] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
According to the neurotrophic hypothesis of major depressive disorder (MDD), impairment in growth factor signaling might be associated with the pathology of this illness. Current evidence demonstrates that impaired neuroplasticity induced by alterations of neurotrophic growth factors and related signaling pathways may be underlying to the pathophysiology of MDD. Brain-derived neurotrophic factor (BDNF) is the most studied neurotrophic factor involved in the neurobiology of MDD. Nevertheless, developing evidence has implicated other neurotrophic factors, including neurotrophin-3 (NT-3), neurotrophin-4 (NT-4), nerve growth factor (NGF), vascular endothelial growth factor (VEGF), insulin-like growth factor (IGF), glial cell-derived neurotrophic factor (GDNF), and fibroblast growth factor (FGF) in the MDD pathophysiology. Here, we summarize the current literature on the involvement of neurotrophic factors and related signaling pathways in the pathophysiology of MDD.
Collapse
|
26
|
Yamagata H, Nakagawa S. Glycosylation and Depression—A Review. TRENDS GLYCOSCI GLYC 2020. [DOI: 10.4052/tigg.2002.1j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Hirotaka Yamagata
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine
| | - Shin Nakagawa
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine
| |
Collapse
|
27
|
Affiliation(s)
- Hirotaka Yamagata
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine
| | - Shin Nakagawa
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine
| |
Collapse
|
28
|
Ly C, Greb AC, Vargas MV, Duim WC, Grodzki ACG, Lein PJ, Olson DE. Transient Stimulation with Psychoplastogens Is Sufficient to Initiate Neuronal Growth. ACS Pharmacol Transl Sci 2020; 4:452-460. [PMID: 33860174 DOI: 10.1021/acsptsci.0c00065] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Indexed: 12/16/2022]
Abstract
Cortical neuron atrophy is a hallmark of depression and includes neurite retraction, dendritic spine loss, and decreased synaptic density. Psychoplastogens, small molecules capable of rapidly promoting cortical neuron growth, have been hypothesized to produce long-lasting positive effects on behavior by rectifying these deleterious structural and functional changes. Here we demonstrate that ketamine and LSD, psychoplastogens from two structurally distinct chemical classes, promote sustained growth of cortical neurons after only short periods of stimulation. Furthermore, we show that psychoplastogen-induced cortical neuron growth can be divided into two distinct epochs: an initial stimulation phase requiring TrkB activation and a growth period involving sustained mTOR and AMPA receptor activation. Our results provide important temporal details concerning the molecular mechanisms by which next-generation antidepressants produce persistent changes in cortical neuron structure, and they suggest that rapidly excreted psychoplastogens might still be effective neurotherapeutics with unique advantages over compounds like ketamine and LSD.
Collapse
Affiliation(s)
- Calvin Ly
- Department of Chemistry, University of California, Davis, One Shields Avenue, Davis, California, Davis 95616, United States
| | - Alexandra C Greb
- Department of Chemistry, University of California, Davis, One Shields Avenue, Davis, California, Davis 95616, United States
| | - Maxemiliano V Vargas
- Neuroscience Graduate Program, University of California, Davis, Davis, California 95618, United States
| | - Whitney C Duim
- Department of Chemistry, University of California, Davis, One Shields Avenue, Davis, California, Davis 95616, United States
| | - Ana Cristina G Grodzki
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, 1089 Veterinary Medicine Drive, Davis, California 95616, United States
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, 1089 Veterinary Medicine Drive, Davis, California 95616, United States
| | - David E Olson
- Department of Chemistry, University of California, Davis, One Shields Avenue, Davis, California, Davis 95616, United States.,Department of Biochemistry & Molecular Medicine, School of Medicine, University of California, Davis, 2700 Stockton Boulevard, Suite 2102, Sacramento, California 95817, United States.,Center for Neuroscience, University of California, Davis, 1544 Newton Ct, Davis, California 95618, United States
| |
Collapse
|
29
|
Anderzhanova E, Hafner K, Genewsky AJ, Soliman A, Pöhlmann ML, Schmidt MV, Blum R, Wotjak CT, Gassen NC. The stress susceptibility factor FKBP51 controls S-ketamine-evoked release of mBDNF in the prefrontal cortex of mice. Neurobiol Stress 2020; 13:100239. [PMID: 33344695 PMCID: PMC7739030 DOI: 10.1016/j.ynstr.2020.100239] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/21/2022] Open
Abstract
We report here the involvement of the stress-responsive glucocorticoid receptor co-chaperone FKBP51 in the mechanism of in vivo secretion of mature BDNF (mBDNF). We used a novel method combining brain microdialysis with a capillary electrophoresis-based immunoassay, to examine mBDNF secretion in the medial prefrontal cortex (mPFC) in vivo in freely moving mice. By combining optogenetic, neurochemical (KCl-evoked depolarization), and transgenic (conditional BDNF knockout mice) means, we have shown that the increase in extracellular mBDNF in vivo is determined by neuronal activity. Withal, mBDNF secretion in the mPFC of mice was stimulated by a systemic administration of S-ketamine (10 or 50 mg/kg) or S-hydroxynorketamine (10 mg/kg). KCl- and S-ketamine-evoked mBDNF secretion was strongly dependent on the expression of FKBP51. Moreover, the inability of S-ketamine to evoke a transient secretion in mBDNF in the mPFC in FKBP51- knockout mice matched the lack of antidepressant-like effect of S-ketamine in the tail suspension test. Our data reveal a critical role of FKBP51 in mBDNF secretion and suggest the involvement of mBDNF in the realization of immediate stress-coping behavior induced by acute S-ketamine.
Collapse
Affiliation(s)
- Elmira Anderzhanova
- Neurohomeostatis Research Group, Clinic of Psychiatry and Psychotherapy University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.,Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinst. 2-10, 80804, Munich, Germany.,BAU International University, Fridon Khalvashi st. 237, Batumi, 6010, Georgia
| | - Kathrin Hafner
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Kraepelinstraße 2-10, 80804, Munich, Germany
| | - Andreas J Genewsky
- Research Group Neuroplasticity, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany.,Department Biology II Cognition and Neural Plasticity, Faculty of Medicine Ludwig-Maximilians Universität München, Großhaderner str. 2, 82152, Planegg-Martinsried, Germany
| | - Azza Soliman
- Research Group Neuroplasticity, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany.,Institute of Human Genetics University Medical Centre, Mainz Langenbeckstr, 155131 Mainz, Germany
| | - Max L Pöhlmann
- Research Group Neurobiology of Stress Resilience, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804, Munich, Germany
| | - Mathias V Schmidt
- Research Group Neurobiology of Stress Resilience, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804, Munich, Germany
| | - Robert Blum
- Institute of Clinical Neurobiology, University Hospital Würzburg, Versbacherstraße 2, 97080, Würzburg, Germany
| | - Carsten T Wotjak
- Research Group Neuroplasticity, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany.,Boehringer Ingelheim Pharma GmbH & Co. KG, Dept. CNS Discovery Research, Birkendorfer Str. 65, 88397, Biberach an der Riß, Germany
| | - Nils C Gassen
- Neurohomeostatis Research Group, Clinic of Psychiatry and Psychotherapy University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.,Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Kraepelinstraße 2-10, 80804, Munich, Germany
| |
Collapse
|
30
|
Tanuma M, Kasai A, Bando K, Kotoku N, Harada K, Minoshima M, Higashino K, Kimishima A, Arai M, Ago Y, Seiriki K, Kikuchi K, Kawata S, Fujita K, Hashimoto H. Direct visualization of an antidepressant analog using surface-enhanced Raman scattering in the brain. JCI Insight 2020; 5:133348. [PMID: 32125287 DOI: 10.1172/jci.insight.133348] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 02/19/2020] [Indexed: 12/18/2022] Open
Abstract
Detailed spatial information of low-molecular weight compound distribution, especially in the brain, is crucial to understanding their mechanism of actions. Imaging techniques that can directly visualize drugs in the brain at a high resolution will complement existing tools for drug distribution analysis. Here, we performed surface-enhanced Raman scattering (SERS) imaging using a bioorthogonal alkyne tag to visualize drugs directly in situ at a high resolution. Focusing on the selective serotonin reuptake inhibitor S-citalopram (S-Cit), which possesses a nitrile group, we substituted an alkynyl group into its structure and synthesized alkynylated S-Cit (Alk-S-Cit). The brain transitivity and the serotonin reuptake inhibition of Alk-S-Cit were not significantly different as compared with S-Cit. Alk-S-Cit was visualized in the coronal mouse brain section using SERS imaging with silver nanoparticles. Furthermore, SERS imaging combined with fluorescence microscopy allowed Alk-S-Cit to be visualized in the adjacent neuronal membranes, as well as in the brain vessel and parenchyma. Therefore, our multimodal imaging technique is an effective method for detecting low-molecular weight compounds in their original tissue environment and can potentially offer additional information regarding the precise spatial distribution of such drugs.
Collapse
Affiliation(s)
- Masato Tanuma
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, and
| | - Atsushi Kasai
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, and
| | - Kazuki Bando
- Department of Applied Physics, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan.,Serendip Research, Osaka, Osaka, Japan
| | - Naoyuki Kotoku
- Chemical Biology Laboratory, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Kazuo Harada
- Department of Legal Medicine, Graduate School of Medicine.,Laboratory of Applied Environmental Biology, Graduate School of Pharmaceutical Sciences
| | | | - Kosuke Higashino
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, and
| | - Atsushi Kimishima
- Laboratory of Natural Products Chemistry, Graduate School of Pharmaceutical Sciences
| | - Masayoshi Arai
- Laboratory of Natural Products Chemistry, Graduate School of Pharmaceutical Sciences
| | - Yukio Ago
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, and.,Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences
| | - Kaoru Seiriki
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, and.,Institute for Transdisciplinary Graduate Degree Programs
| | - Kazuya Kikuchi
- Laboratory of Chemical Biology, Graduate School of Engineering.,Immunology Frontier Research Center, and
| | - Satoshi Kawata
- Department of Applied Physics, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan.,Serendip Research, Osaka, Osaka, Japan
| | - Katsumasa Fujita
- Department of Applied Physics, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan.,Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan.,Advanced Photonics and Biosensing Open Innovation Laboratory, AIST-Osaka University, Suita, Osaka, Japan
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, and.,Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan.,Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka, Japan.,Institute for Datability Science and.,Department of Molecular Pharmaceutical Sciences, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
31
|
Rantamäki T, Kohtala S. Encoding, Consolidation, and Renormalization in Depression: Synaptic Homeostasis, Plasticity, and Sleep Integrate Rapid Antidepressant Effects. Pharmacol Rev 2020; 72:439-465. [DOI: 10.1124/pr.119.018697] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
32
|
Prowse N, Dwyer Z, Thompson A, Fortin T, Elson K, Robeson H, Fenner B, Hayley S. Early life selective knockdown of the TrkB receptor and maternal separation modulates adult stress phenotype. Behav Brain Res 2020; 378:112260. [DOI: 10.1016/j.bbr.2019.112260] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/23/2019] [Accepted: 09/23/2019] [Indexed: 12/21/2022]
|
33
|
Isoflurane produces antidepressant effects inducing BDNF-TrkB signaling in CUMS mice. Psychopharmacology (Berl) 2019; 236:3301-3315. [PMID: 31197433 DOI: 10.1007/s00213-019-05287-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 05/27/2019] [Indexed: 12/21/2022]
Abstract
RATIONALE The volatile anesthetic isoflurane is suggested to produce a rapid and robust antidepressive effect in preliminary clinical trials. Recently, isoflurane was found to activate the tropomyosin receptor kinase B (TrkB) signaling which is the underlying mechanism of the rapid antidepressant ketamine. OBJECTIVE Our study investigated the effect of isoflurane anesthesia on chronic unpredictable mild stressed (CUMS) model in mice and verified the role of brain-derived neurotrophic factor (BDNF)/TrkB/ the mammalian target of rapamycin (mTOR) signaling in the antidepressant effect of isoflurane. METHODS We employed the CUMS model of depression to assess the rapid antidepressant effect of isoflurane by the forced swimming test (FST), the sucrose preference test (SPT), and the novelty suppressed feeding test (NSFT). The protein expression of BDNF and TrkB/protein kinase B (PKB or Akt)/mTOR was determined through Western blot. The dendritic spine density in the hippocampus and medial prefrontal cortex (PFC) was measured by the Golgi staining. RESULTS A brief burst-suppressing isoflurane anesthesia rapidly reversed the behavioral deficits caused by CUMS procedure, normalized the expression of BDNF and further activated the TrkB signaling pathway in CUMS-induced stressed mice in both prefrontal cortex (PFC) and hippocampus (HC). All of those behavioral and proteomic effects were blocked by K252a, a selective receptor inhibitor of TrkB. Isoflurane significantly promoted the formation of dendritic spines in both medial prefrontal cortex (mPFC), CA1, CA3, and DG of the hippocampus. CONCLUSION Our study indicates that isoflurane exerts a rapid antidepressant-like effect in CUMS depression animal model, and the activation of BDNF/TrkB signaling pathway plays an indispensable role in the biological and behavioral antidepressant effects of isoflurane. A single exposure to isoflurane could repair synaptic damage caused by chronic stimulation.
Collapse
|
34
|
Akhter ET, Griffith RW, English AW, Alvarez FJ. Removal of the Potassium Chloride Co-Transporter from the Somatodendritic Membrane of Axotomized Motoneurons Is Independent of BDNF/TrkB Signaling But Is Controlled by Neuromuscular Innervation. eNeuro 2019; 6:ENEURO.0172-19.2019. [PMID: 31541001 PMCID: PMC6795555 DOI: 10.1523/eneuro.0172-19.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/21/2019] [Accepted: 09/15/2019] [Indexed: 01/09/2023] Open
Abstract
The potassium-chloride cotransporter (KCC2) maintains the low intracellular chloride found in mature central neurons and controls the strength and direction of GABA/glycine synapses. We found that following axotomy as a consequence of peripheral nerve injuries (PNIs), KCC2 protein is lost throughout the somatodendritic membrane of axotomized spinal cord motoneurons after downregulation of kcc2 mRNA expression. This large loss likely depolarizes the reversal potential of GABA/glycine synapses, resulting in GABAergic-driven spontaneous activity in spinal motoneurons similar to previous reports in brainstem motoneurons. We hypothesized that the mechanism inducing KCC2 downregulation in spinal motoneurons following peripheral axotomy might be mediated by microglia or motoneuron release of BDNF and TrkB activation as has been reported on spinal cord dorsal horn neurons after nerve injury, motoneurons after spinal cord injury (SCI), and in many other central neurons throughout development or a variety of pathologies. To test this hypothesis, we used genetic approaches to interfere with microglia activation or delete bdnf from specifically microglia or motoneurons, as well as pharmacology (ANA-12) and pharmacogenetics (F616A mice) to block TrkB activation. We show that KCC2 dysregulation in axotomized motoneurons is independent of microglia, BDNF, and TrkB. KCC2 is instead dependent on neuromuscular innervation; KCC2 levels are restored only when motoneurons reinnervate muscle. Thus, downregulation of KCC2 occurs specifically while injured motoneurons are regenerating and might be controlled by target-derived signals. GABAergic and glycinergic synapses might therefore depolarize motoneurons disconnected from their targets and contribute to augment motoneuron activity known to promote motor axon regeneration.
Collapse
Affiliation(s)
- Erica Tracey Akhter
- Departments of Physiology, Emory University, Atlanta, GA 30322
- Cell Biology, Emory University, Atlanta, GA 30322
| | | | | | | |
Collapse
|
35
|
Steinzeig A, Cannarozzo C, Castrén E. Fluoxetine-induced plasticity in the visual cortex outlasts the duration of the naturally occurring critical period. Eur J Neurosci 2019; 50:3663-3673. [PMID: 31299115 PMCID: PMC6899674 DOI: 10.1111/ejn.14512] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 06/18/2019] [Accepted: 07/05/2019] [Indexed: 12/12/2022]
Abstract
Heightened neuronal plasticity expressed during early postnatal life has been thought to permanently decline once critical periods have ended. For example, monocular deprivation is able to shift ocular dominance in the mouse visual cortex during the first months of life, but this effect is lost later in life. However, various treatments, such as the antidepressant fluoxetine, can reactivate a critical period‐like plasticity in the adult brain. When monocular deprivation is supplemented with chronic fluoxetine administration, a major shift in ocular dominance is produced after the critical period has ended. In the current study, we characterized the temporal patterns of fluoxetine‐induced plasticity in the adult mouse visual cortex, using in vivo optical imaging. We found that artificially induced plasticity in ocular dominance extended beyond the duration of the naturally occurring critical period and continued as long as fluoxetine was administered. However, this fluoxetine‐induced plasticity period ended as soon as the drug was not given. These features of antidepressant‐induced plasticity may be useful when designing treatment strategies involving long‐term antidepressant treatment in humans.
Collapse
Affiliation(s)
- Anna Steinzeig
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | | | - Eero Castrén
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
36
|
Culturing primary neurons from rat hippocampus and cortex. Neuronal Signal 2019; 3:NS20180207. [PMID: 32714598 PMCID: PMC7363313 DOI: 10.1042/ns20180207] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/05/2019] [Accepted: 04/15/2019] [Indexed: 01/21/2023] Open
Abstract
Primary neurons from rodent brain hippocampus and cortex have served as important tools in biomedical research over the years. However, protocols for the preparation of primary neurons vary, which often lead to conflicting results. This report provides a robust and reliable protocol for the production of primary neuronal cultures from the cortex and hippocampus with minimal contribution of non-neuronal cells. The neurons were grown in serum-free media and maintained for several weeks without any additional feeder cells. The neuronal cultures maintained according to this protocol differentiate and by 3 weeks develop extensive axonal and dendritic branching. The cultures produced by this method show excellent reproducibility and can be used for histological, molecular and biochemical methods.
Collapse
|
37
|
Abstract
Selective serotonin reuptake inhibitors are among the most prescribed antidepressants. Fluoxetine is the lead molecule which exerts its therapeutic effects, at least in part, by promoting neuroplasticity through increased brain-derived neurotrophic factor (BDNF)/tropomyosin-related receptor kinase B (TrkB) signalling. It is unclear however, to which extent the neuroplastic effects of fluoxetine are solely mediated by the inhibition of the serotonin transporter (5-HTT). To answer this question, the effects of fluoxetine on neuroplasticity were analysed in both wild type (WT) and 5-Htt knock-out (KO) mice. Using Western blotting and RT-qPCR approaches, we showed that fluoxetine 10 µM activated BDNF/TrkB signalling pathways in both CD1 and C57BL/6J mouse primary cortical neurons. Interestingly, effects on BDNF signalling were observed in primary cortical neurons from both 5-Htt WT and KO mice. In addition, a 3-week in vivo fluoxetine treatment (15 mg/kg/d; i.p.) increased the expression of plasticity genes in brains of both 5-Htt WT and KO mice, and tended to equally enhance hippocampal cell proliferation in both genotypes, without reaching significance. Our results further suggest that fluoxetine-induced neuroplasticity does not solely depend on 5-HTT blockade, but might rely, at least in part, on 5-HTT-independent direct activation of TrkB.
Collapse
|
38
|
Diniz CR, Biojone C, Joca SR, Rantamäki T, Castrén E, Guimarães FS, Casarotto PC. Dual mechanism of TRKB activation by anandamide through CB1 and TRPV1 receptors. PeerJ 2019; 7:e6493. [PMID: 30809460 PMCID: PMC6387754 DOI: 10.7717/peerj.6493] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 01/21/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Administration of anandamide (AEA) or 2-arachidonoylglycerol (2AG) induces CB1 coupling and activation of TRKB receptors, regulating the neuronal migration and maturation in the developing cortex. However, at higher concentrations AEA also engages vanilloid receptor TRPV1, usually with opposed consequences on behavior. METHODS AND RESULTS Using primary cell cultures from the cortex of rat embryos (E18) we determined the effects of AEA on phosphorylated TRKB (pTRK). We observed that AEA (at 100 and 200 nM) induced a significant increase in pTRK levels. Such effect of AEA at 100 nM was blocked by pretreatment with the CB1 antagonist AM251 (200 nM) and, at the higher concentration of 200 nM by the TRPV1 antagonist capsazepine (200 nM), but mildly attenuated by AM251. Interestingly, the effect of AEA or capsaicin (a TRPV1 agonist, also at 200 nM) on pTRK was blocked by TRKB.Fc (a soluble form of TRKB able to bind BDNF) or capsazepine, suggesting a mechanism dependent on BDNF release. Using the marble-burying test (MBT) in mice, we observed that the local administration of ACEA (a CB1 agonist) into the prelimbic region of prefrontal cortex (PL-PFC) was sufficient to reduce the burying behavior, while capsaicin or BDNF exerted the opposite effect, increasing the number of buried marbles. In addition, both ACEA and capsaicin effects were blocked by previous administration of k252a (an antagonist of TRK receptors) into PL-PFC. The effect of systemically injected CB1 agonist WIN55,212-2 was blocked by previous administration of k252a. We also observed a partial colocalization of CB1/TRPV1/TRKB in the PL-PFC, and the localization of TRPV1 in CaMK2+ cells. CONCLUSION Taken together, our data indicate that anandamide engages a coordinated activation of TRKB, via CB1 and TRPV1. Thus, acting upon CB1 and TRPV1, AEA could regulate the TRKB-dependent plasticity in both pre- and postsynaptic compartments.
Collapse
Affiliation(s)
- Cassiano R.A.F. Diniz
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Caroline Biojone
- Neuroscience Center—HILIFE, University of Helsinki, Helsinki, Finland
- Department of Physics and Chemistry, Ribeirão Preto School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Samia R.L. Joca
- Department of Physics and Chemistry, Ribeirão Preto School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Clinical Medicine, Translational Neuropsychiatric Unit, Aarhus University, Aarhus, Denmark
- Aarhus Institute of Advanced Studies, Aarhus University, Aarhus, Denmark
| | - Tomi Rantamäki
- Division of Pharmacology and Pharmacotherapeutics, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Eero Castrén
- Neuroscience Center—HILIFE, University of Helsinki, Helsinki, Finland
| | - Francisco S. Guimarães
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Plinio C. Casarotto
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
- Neuroscience Center—HILIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
39
|
Dose-dependent effects of isoflurane on TrkB and GSK3β signaling: Importance of burst suppression pattern. Neurosci Lett 2019; 694:29-33. [DOI: 10.1016/j.neulet.2018.11.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/16/2018] [Accepted: 11/13/2018] [Indexed: 02/06/2023]
|
40
|
TrkB neurotrophin receptor at the core of antidepressant effects, but how? Cell Tissue Res 2019; 377:115-124. [DOI: 10.1007/s00441-018-02985-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/14/2018] [Indexed: 01/19/2023]
|
41
|
Vecchio LM, Meng Y, Xhima K, Lipsman N, Hamani C, Aubert I. The Neuroprotective Effects of Exercise: Maintaining a Healthy Brain Throughout Aging. Brain Plast 2018; 4:17-52. [PMID: 30564545 PMCID: PMC6296262 DOI: 10.3233/bpl-180069] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2018] [Indexed: 02/06/2023] Open
Abstract
Physical activity plays an essential role in maintaining a healthy body, yet it also provides unique benefits for the vascular and cellular systems that sustain a healthy brain. While the benefit of exercise has been observed in humans of all ages, the availability of preclinical models has permitted systematic investigations into the mechanisms by which exercise supports and protects the brain. Over the past twenty-five years, rodent models have shown that increased physical activity elevates neurotrophic factors in the hippocampal and cortical areas, facilitating neurotransmission throughout the brain. Increased physical activity (such as by the voluntary use of a running wheel or regular, timed sessions on a treadmill) also promotes proliferation, maturation and survival of cells in the dentate gyrus, contributing to the process of adult hippocampal neurogenesis. In this way, rodent studies have tremendous value as they demonstrate that an 'active lifestyle' has the capacity to ameliorate a number of age-related changes in the brain, including the decline in adult neurogenesis. Moreover, these studies have shown that greater physical activity may protect the brain health into advanced age through a number of complimentary mechanisms: in addition to upregulating factors in pro-survival neurotrophic pathways and enhancing synaptic plasticity, increased physical activity promotes brain health by supporting the cerebrovasculature, sustaining the integrity of the blood-brain barrier, increasing glymphatic clearance and proteolytic degradation of amyloid beta species, and regulating microglia activation. Collectively, preclinical studies demonstrate that exercise initiates diverse and powerful neuroprotective pathways that may converge to promote continued brain health into old age. This review will draw on both seminal and current literature that highlights mechanisms by which exercise supports the functioning of the brain, and aids in its protection.
Collapse
Affiliation(s)
- Laura M. Vecchio
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada
| | - Ying Meng
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, ON, Canada
- Institute of Medical Sciences, University of Toronto, ON, Canada
| | - Kristiana Xhima
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada
| | - Nir Lipsman
- Institute of Medical Sciences, University of Toronto, ON, Canada
- Physical Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, ON, Canada
| | - Clement Hamani
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, ON, Canada
- Institute of Medical Sciences, University of Toronto, ON, Canada
| | - Isabelle Aubert
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada
| |
Collapse
|
42
|
Di Liberto V, Mudò G, Belluardo N. Crosstalk between receptor tyrosine kinases (RTKs) and G protein-coupled receptors (GPCR) in the brain: Focus on heteroreceptor complexes and related functional neurotrophic effects. Neuropharmacology 2018; 152:67-77. [PMID: 30445101 DOI: 10.1016/j.neuropharm.2018.11.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 11/01/2018] [Accepted: 11/12/2018] [Indexed: 01/11/2023]
Abstract
Neuronal events are regulated by the integration of several complex signaling networks in which G protein-coupled receptors (GPCRs) and receptor tyrosine kinases (RTKs) are considered key players of an intense bidirectional cross-communication in the cell, generating signaling mechanisms that, at the same time, connect and diversify the traditional signal transduction pathways activated by the single receptor. For this receptor-receptor crosstalk, the two classes of receptors form heteroreceptor complexes resulting in RTKs transactivation and in growth-promoting signals. In this review, we describe heteroreceptor complexes between GPCR and RTKs in the central nervous system (CNS) and their functional effects in controlling a variety of neuronal effects, ranging from development, proliferation, differentiation and migration, to survival, repair, synaptic transmission and plasticity. In this interaction, RTKs can also recruit components of the G protein signaling cascade, creating a bidirectional intricate interplay that provides complex control over multiple cellular events. These heteroreceptor complexes, by the integration of different signals, have recently attracted a growing interest as novel molecular target for depressive disorders. This article is part of the Special Issue entitled 'Receptor heteromers and their allosteric receptor-receptor interactions'.
Collapse
Affiliation(s)
- Valentina Di Liberto
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Giuseppa Mudò
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Natale Belluardo
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy.
| |
Collapse
|
43
|
Umemori J, Winkel F, Didio G, Llach Pou M, Castrén E. iPlasticity: Induced juvenile-like plasticity in the adult brain as a mechanism of antidepressants. Psychiatry Clin Neurosci 2018; 72:633-653. [PMID: 29802758 PMCID: PMC6174980 DOI: 10.1111/pcn.12683] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2018] [Indexed: 12/11/2022]
Abstract
The network hypothesis of depression proposes that mood disorders reflect problems in information processing within particular neural networks. Antidepressants (AD), including selective serotonin reuptake inhibitors (SSRI), function by gradually improving information processing within these networks. AD have been shown to induce a state of juvenile-like plasticity comparable to that observed during developmental critical periods: Such critical-period-like plasticity allows brain networks to better adapt to extrinsic and intrinsic signals. We have coined this drug-induced state of juvenile-like plasticity 'iPlasticity.' A combination of iPlasticity induced by chronic SSRI treatment together with training, rehabilitation, or psychotherapy improves symptoms of neuropsychiatric disorders and issues underlying the developmentally or genetically malfunctioning networks. We have proposed that iPlasticity might be a critical component of AD action. We have demonstrated that iPlasticity occurs in the visual cortex, fear erasure network, extinction of aggression caused by social isolation, and spatial reversal memory in rodent models. Chronic SSRI treatment is known to promote neurogenesis and to cause dematuration of granule cells in the dentate gyrus and of interneurons, especially parvalbumin interneurons enwrapped by perineuronal nets in the prefrontal cortex, visual cortex, and amygdala. Brain-derived neurotrophic factor (BDNF), via its receptor tropomyosin kinase receptor B, is involved in the processes of synaptic plasticity, including neurogenesis, neuronal differentiation, weight of synapses, and gene regulation of synaptic formation. BDNF can be activated by both chronic SSRI treatment and neuronal activity. Accordingly, the BDNF/tropomyosin kinase receptor B pathway is critical for iPlasticity, but further analyses will be needed to provide mechanical insight into the processes of iPlasticity.
Collapse
Affiliation(s)
- Juzoh Umemori
- Neuroscience Center, HiLIFEUniversity of HelsinkiHelsinkiFinland
| | - Frederike Winkel
- Neuroscience Center, HiLIFEUniversity of HelsinkiHelsinkiFinland
| | - Giuliano Didio
- Neuroscience Center, HiLIFEUniversity of HelsinkiHelsinkiFinland
| | - Maria Llach Pou
- Neuroscience Center, HiLIFEUniversity of HelsinkiHelsinkiFinland
| | - Eero Castrén
- Neuroscience Center, HiLIFEUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
44
|
Ahmed E, Tawfik MK, Essawy SS, Ahmed AS, Hermans E. Cysteamine Potentiates the Anti-Depressive Effects of Venlafaxine in Corticosterone-Induced Anxiety/Depression Mouse Model: Effect on Brain-Derived Neurotrophic Factor and Tropomyosin-Related Kinase B. EGYPTIAN JOURNAL OF BASIC AND CLINICAL PHARMACOLOGY 2018. [DOI: 10.11131/2018/101383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Affiliation(s)
- Eman Ahmed
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
- Neuropharmacology Group, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Mona K. Tawfik
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Soha S. Essawy
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Amal S. Ahmed
- Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Emmanuel Hermans
- Neuropharmacology Group, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
45
|
Levy MJF, Boulle F, Steinbusch HW, van den Hove DLA, Kenis G, Lanfumey L. Neurotrophic factors and neuroplasticity pathways in the pathophysiology and treatment of depression. Psychopharmacology (Berl) 2018; 235:2195-2220. [PMID: 29961124 PMCID: PMC6061771 DOI: 10.1007/s00213-018-4950-4] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/18/2018] [Indexed: 02/06/2023]
Abstract
Depression is a major health problem with a high prevalence and a heavy socioeconomic burden in western societies. It is associated with atrophy and impaired functioning of cortico-limbic regions involved in mood and emotion regulation. It has been suggested that alterations in neurotrophins underlie impaired neuroplasticity, which may be causally related to the development and course of depression. Accordingly, mounting evidence suggests that antidepressant treatment may exert its beneficial effects by enhancing trophic signaling on neuronal and synaptic plasticity. However, current antidepressants still show a delayed onset of action, as well as lack of efficacy. Hence, a deeper understanding of the molecular and cellular mechanisms involved in the pathophysiology of depression, as well as in the action of antidepressants, might provide further insight to drive the development of novel fast-acting and more effective therapies. Here, we summarize the current literature on the involvement of neurotrophic factors in the pathophysiology and treatment of depression. Further, we advocate that future development of antidepressants should be based on the neurotrophin theory.
Collapse
Affiliation(s)
- Marion J F Levy
- Centre de Psychiatrie et Neurosciences (Inserm U894), Université Paris Descartes, 102-108 rue de la santé, 75014, Paris, France
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands
| | - Fabien Boulle
- Centre de Psychiatrie et Neurosciences (Inserm U894), Université Paris Descartes, 102-108 rue de la santé, 75014, Paris, France
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands
| | - Harry W Steinbusch
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands
| | - Daniël L A van den Hove
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Gunter Kenis
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands
| | - Laurence Lanfumey
- Centre de Psychiatrie et Neurosciences (Inserm U894), Université Paris Descartes, 102-108 rue de la santé, 75014, Paris, France.
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands.
| |
Collapse
|
46
|
Beyond good and evil: A putative continuum-sorting hypothesis for the functional role of proBDNF/BDNF-propeptide/mBDNF in antidepressant treatment. Neurosci Biobehav Rev 2018; 90:70-83. [PMID: 29626490 DOI: 10.1016/j.neubiorev.2018.04.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/13/2018] [Accepted: 04/03/2018] [Indexed: 02/07/2023]
Abstract
Depression and posttraumatic stress disorder are assumed to be maladaptive responses to stress and antidepressants are thought to counteract such responses by increasing BDNF (brain-derived neurotrophic factor) levels. BDNF acts through TrkB (tropomyosin-related receptor kinase B) and plays a central role in neuroplasticity. In contrast, both precursor proBDNF and BDNF propeptide (another metabolic product from proBDNF cleavage) have a high affinity to p75 receptor (p75R) and usually convey apoptosis and neuronal shrinkage. Although BDNF and proBDNF/propeptide apparently act in opposite ways, neuronal turnover and remodeling might be a final common way that both act to promote more effective neuronal networking, avoiding neuronal redundancy and the misleading effects of environmental contingencies. This review aims to provide a brief overview about the BDNF functional role in antidepressant action and about p75R and TrkB signaling to introduce the "continuum-sorting hypothesis." The resulting hypothesis suggests that both BDNF/proBDNF and BDNF/propeptide act as protagonists to fine-tune antidepressant-dependent neuroplasticity in crucial brain structures to modulate behavioral responses to stress.
Collapse
|
47
|
The antidepressant fluoxetine acts on energy balance and leptin sensitivity via BDNF. Sci Rep 2018; 8:1781. [PMID: 29379096 PMCID: PMC5789051 DOI: 10.1038/s41598-018-19886-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 01/08/2018] [Indexed: 01/18/2023] Open
Abstract
Leptin and Brain Derived Neurotrophic Factor (BDNF) pathways are critical players in body weight homeostasis. Noninvasive treatments like environmental stimulation are able to increase response to leptin and induce BDNF expression in the brain. Emerging evidences point to the antidepressant selective serotonin reuptake inhibitor Fluoxetine (FLX) as a drug with effects similar to environmental stimulation. FLX is known to impact on body weight, with mechanisms yet to be elucidated. We herein asked whether FLX affects energy balance, the leptin system and BDNF function. Adult lean male mice chronically treated with FLX showed reduced weight gain, higher energy expenditure, increased sensitivity to acute leptin, increased hypothalamic BDNF expression, associated to changes in white adipose tissue expression typical of “brownization”. In the Ntrk2tm1Ddg/J model, carrying a mutation in the BDNF receptor Tyrosine kinase B (TrkB), these effects are partially or totally reversed. Wild type obese mice treated with FLX showed reduced weight gain, increased energy output, and differently from untreated obese mice, a preserved acute response to leptin in terms of activation of the intracellular leptin transducer STAT3. In conclusion, FLX impacts on energy balance and induces leptin sensitivity and an intact TrkB function is required for these effects to take place.
Collapse
|
48
|
Guo J, Wang H, Tao Q, Sun S, Liu L, Zhang J, Yang D. Antidepressant Imipramine Protects Bupivacaine-Induced Neurotoxicity in Dorsal Root Ganglion Neurons Through Coactivation of TrkA and TrkB. J Cell Biochem 2018; 118:3960-3967. [PMID: 28398601 DOI: 10.1002/jcb.26051] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 04/10/2017] [Indexed: 01/16/2023]
Abstract
In our work, we used an in vitro culture model to investigate whether antidepressant imipramine (Ip) may protect bupivacaine (Bv)-induced neurotoxicity in mouse dorsal root ganglion (DRG). Adult mouse DRG was treated with 5 mM Bv in vitro to induce neurotoxicity. DRG was then pre-treated with Ip, prior to Bv, to examine its effects on protecting Bv-induced DRG apoptosis and neurite degeneration. Ip-induced dynamic changes in Trk receptors, including TrkA/B/C and phosphor (p-)TrkA/B/C, were examined by qPCR and Western blot. TrkA and TrkB were inhibited by siRNAs to further investigate their functional role in Ip- and Bv-treated DRG. Ip protected Bv-induced apoptosis and neurite loss in DRG. Ip did not alter TrkA/B/C expressions, whereas significantly augmented protein productions of p-TrkA and p-TrkB, but not p-TrkC. SiRNA-mediated TrkA or TrkB downregulation inhibited Trk receptors, and reduced p-TrkA and p-TrkB in DRG. TrkA or TrkB downregulation alone had no effect on Ip-induced protection in Bv-injured DRG. However, co-inhibition of TrkA and TrkB significantly ameliorated the protective effect of Ip on Bv-induced apoptosis and neurite loss in DRG. Imipramine protected bupivacaine-induced neurotoxicity in DRG, likely via the co-activation of TrkA and TrkB signaling pathways. J. Cell. Biochem. 118: 3960-3967, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jianrong Guo
- Department of Anesthesiology, Gongli Hospital, Second Military Medical University, Shanghai, 200135, China
| | | | | | | | | | | | - Dawei Yang
- Department of Anesthesiology, Gongli Hospital, Second Military Medical University, Shanghai, 200135, China
| |
Collapse
|
49
|
Gheorghe A, Qiu W, Galea LAM. Hormonal Regulation of Hippocampal Neurogenesis: Implications for Depression and Exercise. Curr Top Behav Neurosci 2018; 43:379-421. [PMID: 30414016 DOI: 10.1007/7854_2018_62] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Adult hippocampal neurogenesis exists in all mammalian species, including humans, and although there has been considerable research investigating the function and regulation of neurogenesis, there remain many open questions surrounding the complexity of this phenomenon. This stems partially from the fact that neurogenesis is a multistage process that involves proliferation, differentiation, migration, survival, and eventual integration of new cells into the existing hippocampal circuitry, each of which can be independently influenced. The function of adult neurogenesis in the hippocampus is related to stress regulation, behavioral efficacy of antidepressants, long-term spatial memory, forgetting, and pattern separation. Steroid hormones influence the regulation of hippocampal neurogenesis, stress regulation, and cognition and differently in males and females. In this chapter, we will briefly tap into the complex network of steroid hormone modulation of neurogenesis in the hippocampus with specific emphasis on stress, testosterone, and estrogen. We examine the possible role of neurogenesis in the etiology of depression and influencing treatment by examining the influence of both pharmacological (selective serotonin reuptake inhibitors, tricyclic antidepressants) treatments and non-pharmacological (exercise) remedies.
Collapse
Affiliation(s)
- Ana Gheorghe
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Wansu Qiu
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada. .,Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada. .,Department of Psychology, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
50
|
Boltaev U, Meyer Y, Tolibzoda F, Jacques T, Gassaway M, Xu Q, Wagner F, Zhang YL, Palmer M, Holson E, Sames D. Multiplex quantitative assays indicate a need for reevaluating reported small-molecule TrkB agonists. Sci Signal 2017; 10:10/493/eaal1670. [PMID: 28831019 DOI: 10.1126/scisignal.aal1670] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) and its receptor, tropomyosin-related kinase B (TrkB), have emerged as key regulators of brain plasticity and represent disease-modifying targets for several brain disorders, including Alzheimer's disease and major depressive disorder. Because of poor pharmacokinetic properties of BDNF, the interest in small-molecule TrkB agonists and modulators is high. Several compounds have been reported to act as TrkB agonists, and their increasing use in various nervous system disorder models creates the perception that these are reliable probes. To examine key pharmacological parameters of these compounds in detail, we have developed and optimized a series of complementary quantitative assays that measure TrkB receptor activation, TrkB-dependent downstream signaling, and gene expression in different cellular contexts. Although BDNF and other neurotrophic factors elicited robust and dose-dependent receptor activation and downstream signaling, we were unable to reproduce these activities using the reported small-molecule TrkB agonists. Our findings indicate that experimental results obtained with these compounds must be carefully interpreted and highlight the challenge of developing reliable pharmacological activators of this key molecular target.
Collapse
Affiliation(s)
- Umed Boltaev
- Department of Chemistry, Columbia University, New York, NY 10027, USA.,NeuroTechnology Center at Columbia University, New York, NY 10027, USA
| | - Yves Meyer
- Department of Chemistry, Columbia University, New York, NY 10027, USA
| | - Farangis Tolibzoda
- Department of Chemistry, Columbia University, New York, NY 10027, USA.,NeuroTechnology Center at Columbia University, New York, NY 10027, USA
| | - Teresa Jacques
- Department of Chemistry, Columbia University, New York, NY 10027, USA
| | - Madalee Gassaway
- Department of Chemistry, Columbia University, New York, NY 10027, USA.,NeuroTechnology Center at Columbia University, New York, NY 10027, USA
| | - Qihong Xu
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, 7 Cambridge Center, Cambridge, MA 02142, USA
| | - Florence Wagner
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, 7 Cambridge Center, Cambridge, MA 02142, USA
| | - Yan-Ling Zhang
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, 7 Cambridge Center, Cambridge, MA 02142, USA
| | - Michelle Palmer
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, 7 Cambridge Center, Cambridge, MA 02142, USA
| | - Edward Holson
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, 7 Cambridge Center, Cambridge, MA 02142, USA
| | - Dalibor Sames
- Department of Chemistry, Columbia University, New York, NY 10027, USA. .,NeuroTechnology Center at Columbia University, New York, NY 10027, USA
| |
Collapse
|