1
|
Wang Y, Deng Y, Feng M, Chen J, Zhong M, Han Z, Zhang Q, Sun Y. Cordycepin Extracted from Cordyceps militaris mitigated CUMS-induced depression of rats via targeting GSK3β/β-catenin signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 340:119249. [PMID: 39689748 DOI: 10.1016/j.jep.2024.119249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/08/2024] [Accepted: 12/13/2024] [Indexed: 12/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cordycepin, the main active component of Cordyceps militaris, exhibits various pharmacological activities, including anti-tumor and antioxidant effects. However, its antidepressant effect and the underlying mechanisms remain unclear. AIM OF REVIEW This study aimed to explore the antidepressant effect of cordycepin and elucidate the potential molecular mechanisms. MATERIALS AND METHODS Chronic unpredictable mild stress (CUMS) rat model was established to assess antidepressant effect of cordycepin. Gas chromatography-mass spectrometry (GC-MS) metabolomics with integrated network pharmacology were used to find differential metabolites in serum, brain, and cerebrospinal fluid of rats and identify potential target by cordycepin. Western blot and Real-time PCR were applied to validate the signaling pathway. RESULTS Cordycepin alleviated CUMS-induced depression-like behaviors by weight gain, sucrose preference increment, immobility time reduction, total travelling distance extension and serum corticosterone levels reduction. Metabolomics showed that cordycepin reversed CUMS-induced metabolic disturbances through alanine and TCA cycle metabolism pathways. Network pharmacology identified GSK3β as a potential target. Cordycepin increased protein levels of p-GSK3β, β-catenin and nuclear β-catenin, and enhanced transcription of downstream genes PKM, LDHA, Cyclin D1 and C-myc in brains of CUMS-induced rats. CONCLUSIONS This study indicated that cordycepin exerted antidepressant effect by modulating GSK3β/β-catenin pathway, suggesting its potential as a candidate agent for depression.
Collapse
Affiliation(s)
- Yupeng Wang
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, the People's Republic of China
| | - Yanhui Deng
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, the People's Republic of China
| | - Mingmei Feng
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, the People's Republic of China
| | - Jiaxi Chen
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, the People's Republic of China
| | - Mengling Zhong
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, the People's Republic of China
| | - Zhipeng Han
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, the People's Republic of China
| | - Qi Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, the People's Republic of China; College of Food Science and Light Industry, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, the People's Republic of China.
| | - Yang Sun
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, the People's Republic of China.
| |
Collapse
|
2
|
Yeh YA, Hsu HC, Lin MC, Chen TS, Lin WC, Huang HM, Lin YW. Electroacupuncture Regulates Cannabinoid Receptor 1 Expression in a Mouse Fibromyalgia Model: Pharmacological and Chemogenetic Modulation. Life (Basel) 2024; 14:1499. [PMID: 39598297 PMCID: PMC11595423 DOI: 10.3390/life14111499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
Fibromyalgia is a chronic illness usually accompanied by long-lasting, general pain throughout the body, often accompanied by anxiety, depression, fatigue, and sleep disruption. Meanwhile, doctors and scientists have not entirely discovered detailed mechanisms; patients always have an exaggerated sensation to pervasive pain without satisfied medical service. Given the lack of knowledge on its underlying mechanism, current treatments aim to provide pain and/or symptom relief. The present study aimed to clarify the role of cannabinoid receptor 1 (CB1) signaling in a mouse fibromyalgia pain model. To develop the mouse fibromyalgia model, mice were subjected to intermittent cold stress (ICS). Our results indicated that mechanical (2.09 ± 0.09 g) and thermal hyperalgesia (4.77 ± 0.29 s), which were evaluated by von Frey and Hargraves' tests, were induced by ICS, suggesting successful modeling. The hurting replies were then provoked by electroacupuncture (EA) but not for sham EA mice. Further, in a Western blot analysis, we found significantly decreased CB1 protein levels in the thalamus, somatosensory cortex, and anterior cingulate cortex. In addition, the levels of pain-related protein kinases and transcription factor were increased. Treatment with EA reliably increased CB1 expression in various brain regions sequentially alleviated by nociceptive mediators. Furthermore, the administration of a CB1 agonist significantly attenuated fibromyalgia pain, reversed EA analgesia by the CB1 antagonist, and further reversed the chemogenetic inhibition of SSC. Our innovative findings evidence the role of CB1 signaling in the interaction of EA and fibromyalgia, suggesting its potential for clinical trials and as a treatment target.
Collapse
Affiliation(s)
- Yu-An Yeh
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung 404328, Taiwan;
| | - Hsin-Cheng Hsu
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 404328, Taiwan;
- Department of Traditional Chinese Medicine, China Medical University Hsinchu Hospital, China Medical University, Hsinchu 302056, Taiwan
| | - Ming-Chia Lin
- Department of Nuclear Medicine, E-DA Hospital, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan;
| | - Tzu-Shan Chen
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung 82445, Taiwan
| | - Wei-Cheng Lin
- Graduate Institute of Sports and Health Management, National Chung Hsing University, Taichung 402202, Taiwan;
| | - Hsiang-Ming Huang
- Department of Neurosurgery, China Medical University Hsinchu Hospital, China Medical University, Hsinchu 302056, Taiwan
| | - Yi-Wen Lin
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung 404328, Taiwan;
- Chinese Medicine Research Center, China Medical University, Taichung 404328, Taiwan
| |
Collapse
|
3
|
Briânis RC, Andreotti JP, Moreira FA, Iglesias LP. Interplay between endocannabinoid and endovanilloid mechanisms in fear conditioning. Acta Neuropsychiatr 2024; 36:255-264. [PMID: 37982167 DOI: 10.1017/neu.2023.54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
OBJECTIVE The transient receptor potential cation channel, subfamily V (vanilloid), member 1 (TRPV1) mediates pain perception to thermal and chemical stimuli in peripheral neurons. The cannabinoid receptor type 1 (CB1), on the other hand, promotes analgesia in both the periphery and the brain. TRPV1 and CB1 have also been implicated in learned fear, which involves the association of a previously neutral stimulus with an aversive event. In this review, we elaborate on the interplay between CB1 receptors and TRPV1 channels in learned fear processing. METHODS We conducted a PubMed search for a narrative review on endocannabinoid and endovanilloid mechanisms on fear conditioning. RESULTS TRPV1 and CB1 receptors are activated by a common endogenous agonist, arachidonoyl ethanolamide (anandamide), Moreover, they are expressed in common neuroanatomical structures and recruit converging cellular pathways, acting in concert to modulate fear learning. However, evidence suggests that TRPV1 exerts a facilitatory role, whereas CB1 restrains fear responses. CONCLUSION TRPV1 and CB1 seem to mediate protective and aversive roles of anandamide, respectively. However, more research is needed to achieve a better understanding of how these receptors interact to modulate fear learning.
Collapse
Affiliation(s)
- Rayssa C Briânis
- Department of Pharmacology, Institute of Biological Sciences; Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Julia P Andreotti
- Department of Pharmacology, Institute of Biological Sciences; Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fabrício A Moreira
- Department of Pharmacology, Institute of Biological Sciences; Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lia P Iglesias
- Department of Pharmacology, Institute of Biological Sciences; Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
4
|
Bellia F, Girella A, Annunzi E, Benatti B, Vismara M, Priori A, Festucci F, Fanti F, Compagnone D, Adriani W, Dell'Osso B, D'Addario C. Selective alterations of endocannabinoid system genes expression in obsessive compulsive disorder. Transl Psychiatry 2024; 14:118. [PMID: 38409080 PMCID: PMC10897168 DOI: 10.1038/s41398-024-02829-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 02/28/2024] Open
Abstract
Obsessive Compulsive Disorder (OCD) is listed as one of the top 10 most disabling neuropsychiatric conditions in the world. The neurobiology of OCD has not been completely understood and efforts are needed in order to develop new treatments. Beside the classical neurotransmitter systems and signalling pathways implicated in OCD, the possible involvement of the endocannabinoid system (ECS) has emerged in pathophysiology of OCD. We report here selective downregulation of the genes coding for enzymes allowing the synthesis of the endocannabinoids. We found reduced DAGLα and NAPE-PLD in blood samples of individuals with OCD (when compared to healthy controls) as well as in the amygdala complex and prefrontal cortex of dopamine transporter (DAT) heterozygous rats, manifesting compulsive behaviours. Also mRNA levels of the genes coding for cannabinoid receptors type 1 and type 2 resulted downregulated, respectively in the rat amygdala and in human blood. Moreover, NAPE-PLD changes in gene expression resulted to be associated with an increase in DNA methylation at gene promoter, and the modulation of this gene in OCD appears to be correlated to the progression of the disease. Finally, the alterations observed in ECS genes expression appears to be correlated with the modulation in oxytocin receptor gene expression, consistently with what recently reported. Overall, we confirm here a role for ECS in OCD at both preclinical and clinical level. Many potential biomarkers are suggested among its components, in particular NAPE-PLD, that might be of help for a prompt and clear diagnosis.
Collapse
Affiliation(s)
- Fabio Bellia
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100, Teramo, Italy
- Center for Advanced Studies and Technology (CAST), University "G. D'Annunzio" of Chieti-Pescara, 66100, Chieti, Italy
| | - Antonio Girella
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100, Teramo, Italy
| | - Eugenia Annunzi
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d' Annunzio" of Chieti-Pescara, 66100, Chieti, Italy
| | - Beatrice Benatti
- Department of Psychiatry, Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, ASST Fatebenefratelli-Sacco, 20019, Milan, Italy
- "Aldo Ravelli" Center for Nanotechnology and Neurostimulation, University of Milan, Milan, Italy
| | - Matteo Vismara
- Department of Psychiatry, Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, ASST Fatebenefratelli-Sacco, 20019, Milan, Italy
| | - Alberto Priori
- Department of Psychiatry, Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, ASST Fatebenefratelli-Sacco, 20019, Milan, Italy
| | - Fabiana Festucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Federico Fanti
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100, Teramo, Italy
| | - Dario Compagnone
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100, Teramo, Italy
| | - Walter Adriani
- Center for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, I-00161, Rome, Italy
| | - Bernardo Dell'Osso
- Department of Psychiatry, Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, ASST Fatebenefratelli-Sacco, 20019, Milan, Italy.
- "Aldo Ravelli" Center for Nanotechnology and Neurostimulation, University of Milan, Milan, Italy.
| | - Claudio D'Addario
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100, Teramo, Italy.
- Department of Clinical Neuroscience, Karolinska Institute, 10316, Stockholm, Sweden.
| |
Collapse
|
5
|
Del Rio R, Serrano RG, Gomez E, Martinez JC, Edward MA, Santos RA, Diaz KS, Cohen-Cory S. Cell-autonomous and differential endocannabinoid signaling impacts the development of presynaptic retinal ganglion cell axon connectivity in vivo. Front Synaptic Neurosci 2023; 15:1176864. [PMID: 37252636 PMCID: PMC10213524 DOI: 10.3389/fnsyn.2023.1176864] [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: 03/01/2023] [Accepted: 04/25/2023] [Indexed: 05/31/2023] Open
Abstract
Cannabis exposure during gestation evokes significant molecular modifications to neurodevelopmental programs leading to neurophysiological and behavioral abnormalities in humans. The main neuronal receptor for Δ9-tetrahydrocannabinol (THC) is the type-1 cannabinoid receptor CB1R, one of the most abundant G-protein-coupled receptors in the nervous system. While THC is the major psychoactive phytocannabinoid, endocannabinoids (eCBs) are the endogenous ligands of CB1R and are known to act as retrograde messengers to modulate synaptic plasticity at different time scales in the adult brain. Accumulating evidence indicates that eCB signaling through activation of CB1R plays a central role in neural development. During development, most CB1R localized to axons of projection neurons, and in mice eCB signaling impacts axon fasciculation. Understanding of eCB-mediated structural plasticity during development, however, requires the identification of the precise spatial and temporal dynamics of CB1R-mediated modifications at the level of individual neurons in the intact brain. Here, the cell-autonomous role of CB1R and the effects of CB1R-mediated eCB signaling were investigated using targeted single-cell knockdown and pharmacologic treatments in Xenopus. We imaged axonal arbors of retinal ganglion cells (RGCs) in real time following downregulation of CB1R via morpholino (MO) knockdown. We also analyzed RGC axons with altered eCB signaling following treatment with URB597, a selective inhibitor of the enzyme that degrades Anandamide (AEA), or JZL184, an inhibitor of the enzyme that blocks 2-Arachidonoylglycerol (2-AG) hydrolysis, at two distinct stages of retinotectal development. Our results demonstrate that CB1R knockdown impacts RGC axon branching at their target and that differential 2-AG and AEA-mediated eCB signaling contributes to presynaptic structural connectivity at the time that axons terminate and when retinotectal synaptic connections are made. Altering CB1R levels through CB1R MO knockdown similarly impacted dendritic morphology of tectal neurons, thus supporting both pre- and postsynaptic cell-autonomous roles for CB1R-mediated eCB signaling.
Collapse
|
6
|
Zhang Y, Wang Q, Zheng Z, Sun Y, Niu Y, Li D, Wang S, Meng W. BDNF enhances electrophysiological activity and excitatory synaptic transmission of RA projection neurons in adult male zebra finches. Brain Res 2023; 1801:148208. [PMID: 36549361 DOI: 10.1016/j.brainres.2022.148208] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/22/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022]
Abstract
The singing of songbirds is a complex vocal behavior. It was reported that brain-derived neurotrophic factor (BDNF), a key neurotrophic factor involved in neuronal survival and activity, plays an important role in regulation of songbirds' song behavior. In all song-related nuclei, the electrophysiological activity of robust nucleus of the arcopallium (RA) in the forebrain of songbirds is directly related to birdsong output. Whether BDNF regulates the electrophysiological activity and synaptic transmission of RA causing the change of song behavior need be further explored. In this study, the effects of BDNF on the electrophysiological activity and excitatory synaptic transmission of RA projection neurons (PNs) in adult male zebra finches were investigated using whole-cell patch clamp recordings in vitro. Our results showed that BDNF increased the firing of evoked action potentials in RA PNs and decreased the membrane input resistance and membrane time constant of RA PNs, indicating that BDNF can promote RA PNs excitability by reducing membrane input resistance and membrane time constant. Meanwhile, BDNF increased the frequency rather than amplitude of miniature excitatory postsynaptic currents (mEPSCs) in RA PNs. Moreover, the effects of BDNF on the excitability, intrinsic membrane properties and mEPSCs of RA PNs were blocked by its receptor TrkB antagonist K252a. These results indicate that BDNF via TrkB enhances the excitability and excitatory synaptic transmission of RA PNs in adult male songbirds through presynaptic mechanisms, suggesting a possible cellular mechanism by which BDNF regulates song behavior.
Collapse
Affiliation(s)
- Yutao Zhang
- Jiangxi Key Laboratory of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Qingqin Wang
- Jiangxi Key Laboratory of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Zijian Zheng
- School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Yalun Sun
- Jiangxi Key Laboratory of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Yali Niu
- School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Dongfeng Li
- School of Life Science, South China Normal University, Guangzhou, China.
| | - Songhua Wang
- Jiangxi Key Laboratory of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang, China.
| | - Wei Meng
- Jiangxi Key Laboratory of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang, China.
| |
Collapse
|
7
|
Martinez Ramirez CE, Ruiz-Pérez G, Stollenwerk TM, Behlke C, Doherty A, Hillard CJ. Endocannabinoid signaling in the central nervous system. Glia 2023; 71:5-35. [PMID: 36308424 PMCID: PMC10167744 DOI: 10.1002/glia.24280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 09/02/2022] [Accepted: 09/29/2022] [Indexed: 11/07/2022]
Abstract
It is hard to overestimate the influence of the endocannabinoid signaling (ECS) system on central nervous system (CNS) function. In the 40 years since cannabinoids were found to trigger specific cell signaling cascades, studies of the ECS system continue to cause amazement, surprise, and confusion! CB1 cannabinoid receptors are expressed widely in the CNS and regulate cell-cell communication via effects on the release of both neurotransmitters and gliotransmitters. CB2 cannabinoid receptors are difficult to detect in the CNS but seem to "punch above their weight" as compounds targeting these receptors have significant effects on inflammatory state and behavior. Positive and negative allosteric modulators for both receptors have been identified and examined in preclinical studies. Concentrations of the endocannabinoid ligands, N-arachidonoylethanolamine and 2-arachidonoylglycerol (2-AG), are regulated by a combination of enzymatic synthesis and degradation and inhibitors of these processes are available and making their way into clinical trials. Importantly, ECS regulates many essential brain functions, including regulation of reward, anxiety, inflammation, motor control, and cellular development. While the field is on the cusp of preclinical discoveries providing impactful clinical and therapeutic insights into many CNS disorders, there is still much to be learned about this remarkable and versatile modulatory system.
Collapse
Affiliation(s)
- César E Martinez Ramirez
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Gonzalo Ruiz-Pérez
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Todd M Stollenwerk
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Christina Behlke
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Ashley Doherty
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Cecilia J Hillard
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
8
|
Van Hook MJ. Brain-derived neurotrophic factor is a regulator of synaptic transmission in the adult visual thalamus. J Neurophysiol 2022; 128:1267-1277. [PMID: 36224192 PMCID: PMC9662800 DOI: 10.1152/jn.00540.2021] [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: 12/16/2021] [Revised: 10/12/2022] [Accepted: 10/12/2022] [Indexed: 11/22/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is an important regulator of circuit development, neuronal survival, and plasticity throughout the nervous system. In the visual system, BDNF is produced by retinal ganglion cells (RGCs) and transported along their axons to central targets. Within the dorsolateral geniculate nucleus (dLGN), a key RGC projection target for conscious vision, the BDNF receptor tropomyosin receptor kinase B (TrkB) is present on RGC axon terminals and postsynaptic thalamocortical (TC) relay neuron dendrites. Based on this, the goal of this study was to determine how BDNF modulates the conveyance of signals through the retinogeniculate (RG) pathway of adult mice. Application of BDNF to dLGN brain slices increased TC neuron spiking evoked by optogenetic stimulation of RGC axons. There was a modest contribution to this effect from a BDNF-dependent enhancement of TC neuron intrinsic excitability including increased input resistance and membrane depolarization. BDNF also increased evoked vesicle release from RGC axon terminals, as evidenced by increased amplitude of evoked excitatory postsynaptic currents (EPSCs), which was blocked by inhibition of TrkB or phospholipase C. High-frequency stimulation revealed that BDNF increased synaptic vesicle pool size, release probability, and replenishment rate. There was no effect of BDNF on EPSC amplitude or short-term plasticity of corticothalamic feedback synapses. Thus, BDNF regulates RG synapses by both presynaptic and postsynaptic mechanisms. These findings suggest that BNDF influences the flow of visual information through the retinogeniculate pathway.NEW & NOTEWORTHY Brain-derived neurotrophic factor (BDNF) is an important regulator of neuronal development and plasticity. In the visual system, BDNF is transported along retinal ganglion cell (RGC) axons to the dorsolateral geniculate nucleus (dLGN), although it is not known how it influences mature dLGN function. Here, BDNF enhanced thalamocortical relay neuron responses to signals arising from RGC axons in the dLGN, pointing toward an important role for BDNF in processing signals en route to the visual cortex.
Collapse
Affiliation(s)
- Matthew J Van Hook
- Truhlsen Eye Institute, Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
9
|
Lemtiri-Chlieh F, Levine ES. 2-AG and anandamide enhance hippocampal long-term potentiation via suppression of inhibition. Front Cell Neurosci 2022; 16:1023541. [PMID: 36212685 PMCID: PMC9534525 DOI: 10.3389/fncel.2022.1023541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
It is widely accepted that exogenous cannabinoids can impair short-term memory and cognition in humans and other animals. This is likely related to the inhibition of long-term potentiation (LTP), a form of synaptic plasticity, by the global and sustained activation of CB1 cannabinoid receptors in the presence of exogenous agonists. Conversely, the temporally and spatially restricted release of endogenous cannabinoid (eCB) ligands may enhance synaptic plasticity in a synapse-specific manner. We examined the role of eCB signaling in LTP by recording field excitatory postsynaptic potentials (fEPSPs) in the CA1 stratum radiatum in hippocampal slices from juvenile mice. LTP was induced either electrically, by theta burst stimulation (TBS), or pharmacologically, by treatment for 15 min with a solution designed to increase intracellular cAMP (chem-LTP). A stable and long-lasting potentiation in fEPSP slope following TBS was significantly reduced by blocking cannabinoid receptor activation with CB1 receptor antagonists. Chem-LTP caused a sustained 2-fold increase in fEPSP slope and was also blocked by CB1 receptor antagonists. TBS-LTP was partially reduced by inhibiting the synthesis of the endogenous ligands 2-arachidonylglycerol (2-AG) and anandamide. A similar effect was observed with chem-LTP. Blocking inhibitory synapses completely prevented the effect of CB1 receptor antagonists or inhibition of eCB synthesis on TBS-LTP and chem-LTP. These results indicate that simultaneous activation of CB1 receptors by 2-AG and anandamide enhances TBS-induced and pharmacologically-induced LTP, and this effect is mediated by the suppression of inhibition at GABAergic synapses.
Collapse
|
10
|
Prenatal exposure to Cannabis smoke induces early and lasting damage to the brain. Neurochem Int 2022; 160:105406. [PMID: 35970295 DOI: 10.1016/j.neuint.2022.105406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 08/02/2022] [Accepted: 08/07/2022] [Indexed: 11/20/2022]
Abstract
Cannabis is the most widely used illegal drug during pregnancy, however, the effects of gestational exposure to Cannabis smoke (CS) on the central nervous system development remain uncharacterised. This study investigates the effects of maternal CS inhalation on brain function in the offspring. Pregnant mice were exposed daily to 5 min of CS during gestational days (GD) 5.5-17.5. On GD 18.5 half of the dams were euthanized for foetus removal. The offspring from the remaining dams were euthanized on postnatal days (PND) 20 and 60 for evaluation. Brain volume, cortex cell number, SOX2, histone-H3, parvalbumin, NeuN, and BDNF immunoreactivity were assessed in all groups. In addition, levels of NeuN, CB1 receptor, and BDNF expression were assessed and cortical primary neurons from rats were treated with Cannabis smoke extract (CSE) for assessment of cell viability. We found that male foetuses from the CS exposed group had decreased brain volume, whereas mice at PND 60 from the exposed group presented with increased brain volume. Olfactory bulb and diencephalon volume were found lower in foetuses exposed to CS. Mice at PND 60 from the exposed group had a smaller volume in the thalamus and hypothalamus while the cerebellum presented with a greater volume. Also, there was an increase in cortical BDNF immunoreactivity in CS exposed mice at PND 60. Protein expression analysis showed an increase in pro-BDNF in foetus brains exposed to CS. Mice at PND 60 presented an increase in mature BDNF in the prefrontal cortex (PFC) in the exposed group and a higher CB1 receptor expression in the PFC. Moreover, hippocampal NeuN expression was higher in adult animals from the exposed group. Lastly, treatment of cortical primary neurons with doses of CSE resulted in decreased cell viability. These findings highlight the potential negative neurodevelopmental outcomes induced by gestational CS exposure.
Collapse
|
11
|
Rapid treatments for depression: Endocannabinoid system as a therapeutic target. Neurosci Biobehav Rev 2022; 137:104635. [PMID: 35351488 DOI: 10.1016/j.neubiorev.2022.104635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 03/19/2022] [Accepted: 03/20/2022] [Indexed: 12/16/2022]
Abstract
Current first-line treatments for major depressive disorder (MDD), i.e., antidepressant drugs and psychotherapy, show delayed onset of therapeutic effect as late as 2-3 weeks or more. In the clinic, the speed of beginning of the actions of antidepressant drugs or other interventions is vital for many reasons. Late-onset means that depression, its related disability, and the potential danger of suicide remain a threat for some patients. There are some rapid-acting antidepressant interventions, such as sleep deprivation, ketamine, acute exercise, which induce a significant response, ranging from a few hours to maximally one week, and most of them share a common characteristic that is the activation of the endocannabinoid (eCB) system. Activation of this system, i.e., augmentation of eCB signaling, appears to have anti-depressant-like actions. This article puts the idea forward that the activation of eCB signaling represents a critical mechanism of rapid-acting therapeutic interventions in MDD, and this system might contribute to the development of novel rapid-acting treatments for MDD.
Collapse
|
12
|
Gao L, Zhang Y, Sterling K, Song W. Brain-derived neurotrophic factor in Alzheimer's disease and its pharmaceutical potential. Transl Neurodegener 2022; 11:4. [PMID: 35090576 PMCID: PMC8796548 DOI: 10.1186/s40035-022-00279-0] [Citation(s) in RCA: 183] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 01/01/2022] [Indexed: 12/14/2022] Open
Abstract
Synaptic abnormalities are a cardinal feature of Alzheimer's disease (AD) that are known to arise as the disease progresses. A growing body of evidence suggests that pathological alterations to neuronal circuits and synapses may provide a mechanistic link between amyloid β (Aβ) and tau pathology and thus may serve as an obligatory relay of the cognitive impairment in AD. Brain-derived neurotrophic factors (BDNFs) play an important role in maintaining synaptic plasticity in learning and memory. Considering AD as a synaptic disorder, BDNF has attracted increasing attention as a potential diagnostic biomarker and a therapeutical molecule for AD. Although depletion of BDNF has been linked with Aβ accumulation, tau phosphorylation, neuroinflammation and neuronal apoptosis, the exact mechanisms underlying the effect of impaired BDNF signaling on AD are still unknown. Here, we present an overview of how BDNF genomic structure is connected to factors that regulate BDNF signaling. We then discuss the role of BDNF in AD and the potential of BDNF-targeting therapeutics for AD.
Collapse
Affiliation(s)
- Lina Gao
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, College of Pharmacy, Jining Medical University, Jining, 272067, Shandong, China
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Yun Zhang
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Keenan Sterling
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Weihong Song
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, College of Pharmacy, Jining Medical University, Jining, 272067, Shandong, China.
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and The Affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, 325001, Zhejiang, China.
| |
Collapse
|
13
|
Babaei P, Azari HB. Exercise Training Improves Memory Performance in Older Adults: A Narrative Review of Evidence and Possible Mechanisms. Front Hum Neurosci 2022; 15:771553. [PMID: 35153701 PMCID: PMC8829997 DOI: 10.3389/fnhum.2021.771553] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/16/2021] [Indexed: 12/15/2022] Open
Abstract
As human life expectancy increases, cognitive decline and memory impairment threaten independence and quality of life. Therefore, finding prevention and treatment strategies for memory impairment is an important health concern. Moreover, a better understanding of the mechanisms involved underlying memory preservation will enable the development of appropriate pharmaceuticals drugs for those who are activity limited. Exercise training as a non-pharmacological tool, has been known to increase the mean lifespan by maintaining general body health and improving the cardiovascular and nervous systems function. Among different exercise training protocols, aerobic exercise has been reported to prevent the progression of memory decline, provided adequate exertion level, duration, and frequency. Mechanisms underlying exercise training effects on memory performance have not been understood yet. Convergent evidence suggest several direct and indirect mechanisms at molecular and supramolecular levels. The supramolecular level includes improvement in blood circulation, synaptic plasticity and neurogenesis which are under controls of complex molecular signaling of neurotransmitters, neurotrophic factors, exerkines, and epigenetics factors. Among these various factors, irisin/BDNF signaling seems to be one of the important mediators of crosstalk between contracted skeletal muscles and the brain during exercise training. This review provides an affordable and effective method to improve cognitive function in old ages, particularly those who are most vulnerable to neurodegenerative disorders.
Collapse
Affiliation(s)
- Parvin Babaei
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Helya Bolouki Azari
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Physiology, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Wang Y, Wu Z, Wang D, Huang C, Xu J, Liu C, Yang C. Muscle-brain communication in pain: The key role of myokines. Brain Res Bull 2021; 179:25-35. [PMID: 34871710 DOI: 10.1016/j.brainresbull.2021.11.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 11/24/2021] [Accepted: 11/28/2021] [Indexed: 12/24/2022]
Abstract
Pain is the most common reason for a physician visit, which accounts for a considerable proportion of the global burden of disease and greatly affects patients' quality of life. Therefore, there is an urgent need to identify new therapeutic targets involved in pain. Exercise-induced hypoalgesia (EIH) is a well known phenomenon observed worldwide. However, the available evidence demonstrates that the mechanisms of EIH remain unclear. One of the most accepted hypotheses has been the activation of several endogenous systems in the brain. Recently, the concept that the muscle acts as a secretory organ has attracted increasing attention. Proteins secreted by the muscle are called myokines, playing a critical role in communicating with other organs, such as the brain. This review will focus on several myokines and discuss their roles in EIH.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zifeng Wu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Di Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chaoli Huang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Jiali Xu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Cunming Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
15
|
Perinatal Fentanyl Exposure Leads to Long-Lasting Impairments in Somatosensory Circuit Function and Behavior. J Neurosci 2021. [PMID: 33853934 DOI: 10.1523/jneurosci.2470-20.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
One consequence of the opioid epidemic are lasting neurodevelopmental sequelae afflicting adolescents exposed to opioids in the womb. A translationally relevant and developmentally accurate preclinical model is needed to understand the behavioral, circuit, network, and molecular abnormalities resulting from this exposure. By employing a novel preclinical model of perinatal fentanyl exposure, our data reveal that fentanyl has several dose-dependent, developmental consequences to somatosensory function and behavior. Newborn male and female mice exhibit signs of withdrawal and sensory-related deficits that extend at least to adolescence. As fentanyl exposure does not affect dams' health or maternal behavior, these effects result from the direct actions of perinatal fentanyl on the pups' developing brain. At adolescence, exposed mice exhibit reduced adaptation to sensory stimuli, and a corresponding impairment in primary somatosensory (S1) function. In vitro electrophysiology demonstrates a long-lasting reduction in S1 synaptic excitation, evidenced by decreases in release probability, NMDA receptor-mediated postsynaptic currents, and frequency of miniature excitatory postsynaptic currents (mEPSCs), as well as increased frequency of miniature inhibitory postsynaptic currents (mIPSCs). In contrast, anterior cingulate cortical neurons exhibit an opposite phenotype, with increased synaptic excitation. Consistent with these changes, electrocorticograms (ECoGs) reveal suppressed ketamine-evoked γ oscillations. Morphologic analysis of S1 pyramidal neurons indicate reduced dendritic complexity, dendritic length, and soma size. Further, exposed mice exhibited abnormal cortical mRNA expression of key receptors involved in synaptic transmission and neuronal growth and development, changes that were consistent with the electrophysiological and morphologic changes. These findings demonstrate the lasting sequelae of perinatal fentanyl exposure on sensory processing and function.SIGNIFICANCE STATEMENT This is the first study to show that exposure to fentanyl in the womb results in behavioral, circuitry, and synaptic effects that last at least to adolescence. We also show, for the first time, that this exposure has different, lasting effects on synapses in different cortical areas.
Collapse
|
16
|
Winters BL, Vaughan CW. Mechanisms of endocannabinoid control of synaptic plasticity. Neuropharmacology 2021; 197:108736. [PMID: 34343612 DOI: 10.1016/j.neuropharm.2021.108736] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 01/13/2023]
Abstract
The endogenous cannabinoid transmitter system regulates synaptic transmission throughout the nervous system. Unlike conventional transmitters, specific stimuli induce synthesis of endocannabinoids (eCBs) in the postsynaptic neuron, and these travel backwards to modulate presynaptic inputs. In doing so, eCBs can induce short-term changes in synaptic strength and longer-term plasticity. While this eCB regulation is near ubiquitous, it displays major regional and synapse specific variations with different synapse specific forms of short-versus long-term plasticity throughout the brain. These differences are due to the plethora of pre- and postsynaptic mechanisms which have been implicated in eCB signalling, the intricacies of which are only just being realised. In this review, we shall describe the current understanding and highlight new advances in this area, with a focus on the retrograde action of eCBs at CB1 receptors (CB1Rs).
Collapse
Affiliation(s)
- Bryony Laura Winters
- Pain Management Research Institute, Kolling Institute of Medical Research, Northern Clinical School, University of Sydney at Royal North Shore Hospital, NSW, Australia.
| | - Christopher Walter Vaughan
- Pain Management Research Institute, Kolling Institute of Medical Research, Northern Clinical School, University of Sydney at Royal North Shore Hospital, NSW, Australia
| |
Collapse
|
17
|
Alipio JB, Haga C, Fox ME, Arakawa K, Balaji R, Cramer N, Lobo MK, Keller A. Perinatal Fentanyl Exposure Leads to Long-Lasting Impairments in Somatosensory Circuit Function and Behavior. J Neurosci 2021; 41:3400-3417. [PMID: 33853934 PMCID: PMC8051687 DOI: 10.1523/jneurosci.2470-20.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/07/2020] [Accepted: 12/30/2020] [Indexed: 11/21/2022] Open
Abstract
One consequence of the opioid epidemic are lasting neurodevelopmental sequelae afflicting adolescents exposed to opioids in the womb. A translationally relevant and developmentally accurate preclinical model is needed to understand the behavioral, circuit, network, and molecular abnormalities resulting from this exposure. By employing a novel preclinical model of perinatal fentanyl exposure, our data reveal that fentanyl has several dose-dependent, developmental consequences to somatosensory function and behavior. Newborn male and female mice exhibit signs of withdrawal and sensory-related deficits that extend at least to adolescence. As fentanyl exposure does not affect dams' health or maternal behavior, these effects result from the direct actions of perinatal fentanyl on the pups' developing brain. At adolescence, exposed mice exhibit reduced adaptation to sensory stimuli, and a corresponding impairment in primary somatosensory (S1) function. In vitro electrophysiology demonstrates a long-lasting reduction in S1 synaptic excitation, evidenced by decreases in release probability, NMDA receptor-mediated postsynaptic currents, and frequency of miniature excitatory postsynaptic currents (mEPSCs), as well as increased frequency of miniature inhibitory postsynaptic currents (mIPSCs). In contrast, anterior cingulate cortical neurons exhibit an opposite phenotype, with increased synaptic excitation. Consistent with these changes, electrocorticograms (ECoGs) reveal suppressed ketamine-evoked γ oscillations. Morphologic analysis of S1 pyramidal neurons indicate reduced dendritic complexity, dendritic length, and soma size. Further, exposed mice exhibited abnormal cortical mRNA expression of key receptors involved in synaptic transmission and neuronal growth and development, changes that were consistent with the electrophysiological and morphologic changes. These findings demonstrate the lasting sequelae of perinatal fentanyl exposure on sensory processing and function.SIGNIFICANCE STATEMENT This is the first study to show that exposure to fentanyl in the womb results in behavioral, circuitry, and synaptic effects that last at least to adolescence. We also show, for the first time, that this exposure has different, lasting effects on synapses in different cortical areas.
Collapse
Affiliation(s)
- Jason B Alipio
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Catherine Haga
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Megan E Fox
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Keiko Arakawa
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Rakshita Balaji
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Nathan Cramer
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Mary Kay Lobo
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Asaf Keller
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201
| |
Collapse
|
18
|
Durieux LJA, Gilissen SRJ, Arckens L. Endocannabinoids and cortical plasticity: CB1R as a possible regulator of the excitation/inhibition balance in health and disease. Eur J Neurosci 2021; 55:971-988. [PMID: 33427341 DOI: 10.1111/ejn.15110] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/27/2022]
Abstract
The endocannabinoid system has been linked to neurological disorders in which the excitation inhibition (E/I) balance in the neocortex is dysregulated, such as schizophrenia. The main endocannabinoid receptor type 1 of the central nervous system-CB1R-is expressed on different cell types, that when activated, modulate the cortical E/I balance. Here we review how CB1R signalling contributes to phases of heightened plasticity of the neocortex. We review the major role of the CB1R in cortical plasticity throughout life, including the early life sensory critical periods, the later maturation phase of the association cortex in adolescence, and the adult phase of sensory deprivation-induced cortical plasticity. Endocannabinoid-mediated long-term potentiation and depression of synapse strength fine-tune the E/I balance in visual, somatosensory and association areas. We emphasize how a distinct set of key endocannabinoid-regulated elements such as GABA and glutamate release, basket parvalbumin interneurons, somatostatin interneurons and astrocytes, are essential for normal cortical plasticity and dysregulated in schizophrenia. Even though a lot of data has been gathered, mechanistic knowledge about the exact CB1R-based modulation of excitation and/or inhibition is still lacking depending on cortical area and maturation phase in life. We emphasize the importance of creating such detailed knowledge for a better comprehension of what underlies the dysregulation of the neocortex in schizophrenic patients in adulthood. We propose that taking age, brain area and cell type into consideration when modulating the cortical E/I imbalance via cannabinoid-based pharmacology may pave the way for better patient care.
Collapse
Affiliation(s)
- Lucas J A Durieux
- KU Leuven, Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, Leuven, Belgium.,Leuven Brain Institute, Leuven, Belgium
| | - Sara R J Gilissen
- KU Leuven, Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, Leuven, Belgium.,Leuven Brain Institute, Leuven, Belgium
| | - Lutgarde Arckens
- KU Leuven, Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, Leuven, Belgium.,Leuven Brain Institute, Leuven, Belgium
| |
Collapse
|
19
|
Wang X, Jiao X, Xu M, Wang B, Li J, Yang F, Zhang L, Xu L, Yu X. Effects of circulating vitamin D concentrations on emotion, behavior and attention: A cross-sectional study in preschool children with follow-up behavior experiments in juvenile mice. J Affect Disord 2020; 275:290-298. [PMID: 32734921 DOI: 10.1016/j.jad.2020.06.043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/31/2020] [Accepted: 06/16/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Although few studies show that vitamin D (VitD) deficiency has a negative effect on children's emotion and behavior, the effects of the excessive VitD and the appropriate 25(OH)D concentration have never been reported. We investigated the effect of the deficient and excessive VitD on emotion, behavior and attention. METHODS 351 preschool children in a multicenter study in Shanghai, China that had serum 25(OH)D measurements and emotion, behavior and attention measures were included in the analyses. In animal experiments, C57 mice were randomly assigned to three groups (n = 8): control (C) group, VitD deficiency (VDD) group, and VitD overdose (VDO) group. The emotion, behavior and attention of juvenile mice were evaluated through the behavioral experiments. RESULTS There was an "U" relationship between serum 25(OH)D concentration and emotion, behavior and attention. Compared with 20-40 ng/mL group, the odds ratios (ORs) were 1.5 (1.0, 4.8) for emotional problem, 3.8 (1.2, 12.1) for conduct problem and 1.8 (1.1, 5.7) for inattention in <20 ng/mL group. Meanwhile, compared with 20-40 ng/mL group, ORs were 9.5 (2.9, 31.4) for impulsive hyperactivity, and 3.9 (1.2, 12.9) for conduct problem in >40 ng/mL group. Consistent with the results in children, animal experiments showed that the attention level decreased in VDD group, while the anxiety level, hyperactive level and aggressiveness in VDD group and VDO group were significantly increased, respectively. LIMITATIONS 25(OH)D measurements were only available in one season. CONCLUSION The deficient and excessive VitD status both adversely affected children's emotion, behavior and attention.
Collapse
Affiliation(s)
- Xirui Wang
- Department of Developmental and Behavioral Pediatrics, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Pudong, Shanghai 200127, China
| | - Xianting Jiao
- MOE-Shanghai Key Lab of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Yangpu, Shanghai 200092, China
| | - Mingqing Xu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 South Wanping Road, Xuhui, Shanghai 200030, China
| | - Bin Wang
- Department of Developmental and Behavioral Pediatrics, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Pudong, Shanghai 200127, China
| | - Juan Li
- Department of Developmental and Behavioral Pediatrics, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Pudong, Shanghai 200127, China
| | - Fan Yang
- Department of Developmental and Behavioral Pediatrics, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Pudong, Shanghai 200127, China
| | - Lishan Zhang
- Department of Child Health Care, Shanghai Pudong Maternal and Child Health Care Institution, Pudong, Shanghai 201399, China
| | - Lei Xu
- Department of Child Health Care, Shanghai Pudong Maternal and Child Health Care Institution, Pudong, Shanghai 201399, China
| | - Xiaodan Yu
- Department of Developmental and Behavioral Pediatrics, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Pudong, Shanghai 200127, China; MOE-Shanghai Key Lab of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Yangpu, Shanghai 200092, China.
| |
Collapse
|
20
|
BDNF Activates Postsynaptic TrkB Receptors to Induce Endocannabinoid Release and Inhibit Presynaptic Calcium Influx at a Calyx-Type Synapse. J Neurosci 2020; 40:8070-8087. [PMID: 32948677 PMCID: PMC7574661 DOI: 10.1523/jneurosci.2838-19.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 09/03/2020] [Accepted: 09/13/2020] [Indexed: 12/26/2022] Open
Abstract
Brain-derived neurotropic factor (BDNF) has been shown to play critical roles in neural development, plasticity, and neurodegenerative diseases. The main function of BDNF in the brain is widely accepted to be synaptic regulation. However, how BDNF modulates synaptic transmission, especially the underlying signaling cascades between presynaptic and postsynaptic neurons, remains controversial. Brain-derived neurotropic factor (BDNF) has been shown to play critical roles in neural development, plasticity, and neurodegenerative diseases. The main function of BDNF in the brain is widely accepted to be synaptic regulation. However, how BDNF modulates synaptic transmission, especially the underlying signaling cascades between presynaptic and postsynaptic neurons, remains controversial. In the present study, we investigated the actions of BDNF at rat calyx-type synapses of either sex by measuring the excitatory postsynaptic current (EPSC) and presynaptic calcium current and capacitance changes. We found that BDNF inhibits the EPSC, presynaptic calcium influx, and exocytosis/endocytosis via activation of the presynaptic cannabinoid Type 1 receptors (CB1Rs). Inhibition of the CB1Rs abolished the BDNF-induced presynaptic inhibition, whereas CB1R agonist mimicked the effect of BDNF. Exploring the underlying signaling cascade, we found that BDNF specifically activates the postsynaptic TrkB receptors, inducing the release of endocannabinoids via the PLCγ/DGL pathway and retrogradely activating presynaptic CB1Rs. We also reported the involvement of AC/PKA in modulating vesicle endocytosis, which may account for the BDNF-induced calcium-dependent and -independent regulation of endocytosis. Thus, our study provides new insights into the BDNF/endocannabinoid-associated modulation of neurotransmission in physiological and pathologic processes. SIGNIFICANCE STATEMENT BDNF plays critical roles in the modulation of synaptic strength. However, how BDNF regulates synaptic transmission and its underlying signaling cascade(s) remains elusive. By measuring EPSC and the presynaptic calcium current and capacitance changes at rat calyces, we found that BDNF inhibits synaptic transmission via BDNF-TrkB-eCB signaling pathway. Activation of postsynaptic TrkB receptors induces endocannabinoid release via the PLCγ/DGL pathway, retrogradely activating the presynaptic CB1Rs, inhibiting the AC/PKA, and suppressing calcium influx. Our findings provide a comprehensive understanding of BDNF/endocannabinoid-associated modulation of neuronal activities.
Collapse
|
21
|
Gangarossa G, Perez S, Dembitskaya Y, Prokin I, Berry H, Venance L. BDNF Controls Bidirectional Endocannabinoid Plasticity at Corticostriatal Synapses. Cereb Cortex 2019; 30:197-214. [DOI: 10.1093/cercor/bhz081] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/19/2019] [Accepted: 03/20/2019] [Indexed: 12/12/2022] Open
Abstract
AbstractThe dorsal striatum exhibits bidirectional corticostriatal synaptic plasticity, NMDAR and endocannabinoids (eCB) mediated, necessary for the encoding of procedural learning. Therefore, characterizing factors controlling corticostriatal plasticity is of crucial importance. Brain-derived neurotrophic factor (BDNF) and its receptor, the tropomyosine receptor kinase-B (TrkB), shape striatal functions, and their dysfunction deeply affects basal ganglia. BDNF/TrkB signaling controls NMDAR plasticity in various brain structures including the striatum. However, despite cross-talk between BDNF and eCBs, the role of BDNF in eCB plasticity remains unknown. Here, we show that BDNF/TrkB signaling promotes eCB-plasticity (LTD and LTP) induced by rate-based (low-frequency stimulation) or spike-timing–based (spike-timing–dependent plasticity, STDP) paradigm in striatum. We show that TrkB activation is required for the expression and the scaling of both eCB-LTD and eCB-LTP. Using 2-photon imaging of dendritic spines combined with patch-clamp recordings, we show that TrkB activation prolongs intracellular calcium transients, thus increasing eCB synthesis and release. We provide a mathematical model for the dynamics of the signaling pathways involved in corticostriatal plasticity. Finally, we show that TrkB activation enlarges the domain of expression of eCB-STDP. Our results reveal a novel role for BDNF/TrkB signaling in governing eCB-plasticity expression in striatum and thus the engram of procedural learning.
Collapse
Affiliation(s)
- Giuseppe Gangarossa
- Center for Interdisciplinary Research in Biology, College de France, Centre National de la Recherche Scientifique (CNRS) UMR, Institut National de la Santé et de la Recherche (INSERM), Paris Sciences et Lettres (PSL) Research University, Paris, France
| | - Sylvie Perez
- Center for Interdisciplinary Research in Biology, College de France, Centre National de la Recherche Scientifique (CNRS) UMR, Institut National de la Santé et de la Recherche (INSERM), Paris Sciences et Lettres (PSL) Research University, Paris, France
| | - Yulia Dembitskaya
- Center for Interdisciplinary Research in Biology, College de France, Centre National de la Recherche Scientifique (CNRS) UMR, Institut National de la Santé et de la Recherche (INSERM), Paris Sciences et Lettres (PSL) Research University, Paris, France
| | - Ilya Prokin
- INRIA, Villeurbanne, France
- University of Lyon, LIRIS UMR, Villeurbanne, France
| | - Hugues Berry
- INRIA, Villeurbanne, France
- University of Lyon, LIRIS UMR, Villeurbanne, France
| | - Laurent Venance
- Center for Interdisciplinary Research in Biology, College de France, Centre National de la Recherche Scientifique (CNRS) UMR, Institut National de la Santé et de la Recherche (INSERM), Paris Sciences et Lettres (PSL) Research University, Paris, France
| |
Collapse
|
22
|
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: 2.0] [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
|
23
|
Ferreira FF, Ribeiro FF, Rodrigues RS, Sebastião AM, Xapelli S. Brain-Derived Neurotrophic Factor (BDNF) Role in Cannabinoid-Mediated Neurogenesis. Front Cell Neurosci 2018; 12:441. [PMID: 30546297 PMCID: PMC6279918 DOI: 10.3389/fncel.2018.00441] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/05/2018] [Indexed: 12/13/2022] Open
Abstract
The adult mammalian brain can produce new neurons in a process called adult neurogenesis, which occurs mainly in the subventricular zone (SVZ) and in the hippocampal dentate gyrus (DG). Brain-derived neurotrophic factor (BDNF) signaling and cannabinoid type 1 and 2 receptors (CB1R and CB2R) have been shown to independently modulate neurogenesis, but how they may interact is unknown. We now used SVZ and DG neurosphere cultures from early (P1-3) postnatal rats to study the CB1R and CB2R crosstalk with BDNF in modulating neurogenesis. BDNF promoted an increase in SVZ and DG stemness and cell proliferation, an effect blocked by a CB2R selective antagonist. CB2R selective activation promoted an increase in DG multipotency, which was inhibited by the presence of a BDNF scavenger. CB1R activation induced an increase in SVZ and DG cell proliferation, being both effects dependent on BDNF. Furthermore, SVZ and DG neuronal differentiation was facilitated by CB1R and/or CB2R activation and this effect was blocked by sequestering endogenous BDNF. Conversely, BDNF promoted neuronal differentiation, an effect abrogated in SVZ cells by CB1R or CB2R blockade while in DG cells was inhibited by CB2R blockade. We conclude that endogenous BDNF is crucial for the cannabinoid-mediated effects on SVZ and DG neurogenesis. On the other hand, cannabinoid receptor signaling is also determinant for BDNF actions upon neurogenesis. These findings provide support for an interaction between BDNF and endocannabinoid signaling to control neurogenesis at distinct levels, further contributing to highlight novel mechanisms in the emerging field of brain repair.
Collapse
Affiliation(s)
- Filipa Fiel Ferreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Filipa F Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Rui S Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Maria Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
24
|
Selvam R, Yeh ML, Levine ES. Endogenous cannabinoids mediate the effect of BDNF at CA1 inhibitory synapses in the hippocampus. Synapse 2018; 73:e22075. [PMID: 30334291 PMCID: PMC6470051 DOI: 10.1002/syn.22075] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/20/2018] [Accepted: 10/12/2018] [Indexed: 12/14/2022]
Abstract
Brain-derived neurotrophic factor (BDNF), traditionally known for promoting neuronal growth and development, is also a modulator of synaptic transmission. In addition to the well-characterized effects at excitatory synapses, BDNF has been shown to acutely suppress inhibitory neurotransmission; however, the underlying mechanisms are unclear. We have previously shown that at inhibitory synapses in layer 2/3 of the somatosensory cortex, BDNF induces the mobilization of endogenous cannabinoids (eCBs) that act retrogradely to suppress GABA release. Here, we hypothesized that in the hippocampus, BDNF acts similarly via eCB signaling to suppress GABAergic transmission. We found that the acute application of BDNF reduced the spontaneous inhibitory postsynaptic currents (sIPSCs) via postsynaptic TrkB receptor activation. The suppressive effects of BDNF required eCB signaling, as this effect on sIPSCs was prevented by a CB1 receptor antagonist. Further, blocking the postsynaptic eCB release prevented the effect of BDNF, whereas eCB reuptake inhibition enhanced the effect of BDNF. These results suggest that BDNF triggers the postsynaptic release of eCBs. To identify the specific eCB release by BDNF, we tested the effects of disrupting the synthesis or degradation of 2-arachidonoylcglycerol (2-AG). Blocking 2-AG synthesis prevented the effect of BDNF and blocking 2-AG degradation enhanced the effect of BDNF. However, there was no change in the effect of BDNF when anandamide degradation was blocked. Collectively, these results suggest that in the hippocampus, BDNF-TrkB signaling induces the postsynaptic release of the endogenous cannabinoid 2-AG, which acts retrogradely on the presynaptic CB1 receptors to suppress GABA release.
Collapse
Affiliation(s)
- Rajamani Selvam
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut
| | - Mason L Yeh
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut
| | - Eric S Levine
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut
| |
Collapse
|
25
|
Augustin SM, Lovinger DM. Functional Relevance of Endocannabinoid-Dependent Synaptic Plasticity in the Central Nervous System. ACS Chem Neurosci 2018; 9:2146-2161. [PMID: 29400439 DOI: 10.1021/acschemneuro.7b00508] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The endocannabinoid (eCB) signaling system plays a key role in short-term and long-term synaptic plasticity in brain regions involved in various neural functions ranging from action selection to appetite control. This review will explore the role of eCBs in shaping neural circuit function to regulate behaviors. In particular, we will discuss the behavioral consequences of eCB mediated long-term synaptic plasticity in different brain regions. This review brings together evidence from in vitro and ex vivo studies and points out the need for more in vivo studies.
Collapse
Affiliation(s)
- Shana M. Augustin
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland 20852, United States
| | - David M. Lovinger
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland 20852, United States
| |
Collapse
|
26
|
Synaptic and circuit development of the primary sensory cortex. Exp Mol Med 2018; 50:1-9. [PMID: 29628505 PMCID: PMC5938038 DOI: 10.1038/s12276-018-0029-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 12/06/2017] [Indexed: 01/06/2023] Open
Abstract
Animals, including humans, optimize their primary sensory cortex through the use of input signals, which allow them to adapt to the external environment and survive. The time window at the beginning of life in which external input signals are connected sensitively and strongly to neural circuit optimization is called the critical period. The critical period has attracted the attention of many neuroscientists due to the rapid activity-/experience-dependent circuit development that occurs, which is clearly differentiated from other developmental time periods and brain areas. This process involves various types of GABAergic inhibitory neurons, the extracellular matrix, neuromodulators, transcription factors, and neurodevelopmental factors. In this review, I discuss recent progress regarding the biological nature of the critical period that contribute to a better understanding of brain development.
Collapse
|
27
|
Gandhi K, Li C, German N, Skobowiat C, Carrillo M, Kallem RR, Larumbe E, Martinez S, Chuecos M, Ventolini G, Nathanielsz P, Schlabritz-Loutsevitch N. Effect of maternal high-fat diet on key components of the placental and hepatic endocannabinoid system. Am J Physiol Endocrinol Metab 2018; 314:E322-E333. [PMID: 29138223 PMCID: PMC5966752 DOI: 10.1152/ajpendo.00119.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 10/31/2017] [Accepted: 11/01/2017] [Indexed: 02/06/2023]
Abstract
Maternal obesity in pregnancy has been linked to a spectrum of adverse developmental changes. Involvement of eCBs in obesity is well characterized. However, information regarding eCB physiology in obesity associated with pregnancy is sparse. This study evaluated fetomaternal hepatic, systemic, and placental eCB molecular changes in response to maternal consumption of a HFD. From ≥9 mo before conception, nonpregnant baboons ( Papio spp.) were fed a diet of either 45 (HFD; n = 11) or 12% fat or a control diet (CTR; n = 11), and dietary intervention continued through pregnancy. Maternal and fetal venous plasma samples were evaluated using liquid chromatography-mass spectrometry to quantify AEA and 2-AG. Placental, maternal and fetal hepatic tissues were analyzed using RT-PCR, Western blot, and immunohistochemistry. mRNA and protein expression of endocannabinoid receptors (CB1R and CB2R), FAAH, DAGL, MAGL, and COX-2 were determined. Statistical analyses were performed with the nonparametric Scheirer-Ray-Hare extension of the Kruskal-Wallis test to analyze the effects of diet (HFD vs. CTR), fetal sex (male vs. female), and the diet × sex interaction. Fetal weight was influenced by fetal sex but not by maternal diet. The increase in maternal weight in animals fed the HFD vs. the CTR diet approached significance ( P = 0.055). Maternal circulating 2-AG concentrations increased, and fetal circulating concentrations decreased in the HFD group, independently of fetal sex. CB1R receptor expression was detected in syncytiotrophoblasts (HFD) and the fetal endothelium (CTR and HFD). Placental CB2R protein expression was higher in males and lower in female fetuses in the HFD group. Fetal hepatic CB2R, FAAH, COX-2 (for both fetal sexes), and DAGLα (in male fetuses) protein expression decreased in the HFD group compared with the CTR group. We conclude that consumption of a HFD during pregnancy results in fetal systemic 2-AG and hepatic eCB deficiency.
Collapse
Affiliation(s)
- Kushal Gandhi
- School of Medicine, Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center at the Permian Basin , Odessa, Texas
| | - Cun Li
- Department of Animal Science, University of Wyoming , Laramie, Wyoming
- Texas Biomedical Research Institute and Southwest National Primate Research Center , San Antonio, Texas
| | - Nadezhda German
- School of Pharmacy, Texas Tech University Health Sciences Center , Amarillo, Texas
| | | | - Maira Carrillo
- School of Medicine, Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center at the Permian Basin , Odessa, Texas
| | - Raja Reddy Kallem
- School of Pharmacy, Texas Tech University Health Sciences Center , Amarillo, Texas
| | - Eneko Larumbe
- Clinical Research Institute, Texas Tech University Health Sciences , Lubbock, Texas
| | - Stacy Martinez
- School of Medicine, Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center at the Permian Basin , Odessa, Texas
| | - Marcel Chuecos
- School of Medicine, Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center at the Permian Basin , Odessa, Texas
| | - Gary Ventolini
- School of Medicine, Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center at the Permian Basin , Odessa, Texas
| | - Peter Nathanielsz
- Department of Animal Science, University of Wyoming , Laramie, Wyoming
- Texas Biomedical Research Institute and Southwest National Primate Research Center , San Antonio, Texas
| | - Natalia Schlabritz-Loutsevitch
- School of Medicine, Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center at the Permian Basin , Odessa, Texas
| |
Collapse
|
28
|
Llorente-Ovejero A, Manuel I, Giralt MT, Rodríguez-Puertas R. Increase in cortical endocannabinoid signaling in a rat model of basal forebrain cholinergic dysfunction. Neuroscience 2017; 362:206-218. [PMID: 28827178 DOI: 10.1016/j.neuroscience.2017.08.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 07/20/2017] [Accepted: 08/03/2017] [Indexed: 12/24/2022]
Abstract
The basal forebrain cholinergic pathways progressively degenerate during the progression of Alzheimer's disease, leading to an irreversible impairment of memory and thinking skills. The stereotaxic lesion with 192IgG-saporin in the rat brain has been used to eliminate basal forebrain cholinergic neurons and is aimed at emulating the cognitive damage described in this disease in order to explore its effects on behavior and on neurotransmission. Learning and memory processes that are controlled by cholinergic neurotransmission are also modulated by the endocannabinoid (eCB) system. The objective of the present study is to evaluate the eCB signaling in relation to the memory impairment induced in adult rats following a specific cholinergic lesion of the basal forebrain. Therefore, CB1 receptor-mediated signaling was analyzed using receptor and functional autoradiography, and cellular distribution by immunofluorescence. The passive avoidance test and histochemical data revealed a relationship between impaired behavioral responses and a loss of approximately 75% of cholinergic neurons in the nucleus basalis magnocellularis (NBM), accompanied by cortical cholinergic denervation. The decrease in CB1 receptor density observed in the hippocampus, together with hyperactivity of eCB signaling in the NBM and cortex, suggest an interaction between the eCB and cholinergic systems. Moreover, following basal forebrain cholinergic denervation, the presynaptic GABAergic immunoreactivity was reduced in cortical areas. In conclusion, CB1 receptors present in presynaptic GABAergic terminals in the hippocampus are down regulated, but not those in cortical glutamatergic synapses.
Collapse
Affiliation(s)
- Alberto Llorente-Ovejero
- Department of Pharmacology, Faculty of Medicine and Nursing. University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Iván Manuel
- Department of Pharmacology, Faculty of Medicine and Nursing. University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Maria Teresa Giralt
- Department of Pharmacology, Faculty of Medicine and Nursing. University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Rafael Rodríguez-Puertas
- Department of Pharmacology, Faculty of Medicine and Nursing. University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain.
| |
Collapse
|
29
|
Abstract
Autism spectrum disorders (ASDs) are diagnosed on the basis of three behavioral features, namely, (1) deficits in social communication, (2) absence or delay in language and (3) stereotypy. The consensus regarding the neurological pathogenesis of ASDs is aberrant synaptogenesis and synapse function. Further, it is now widely accepted that ASD is neurodevelopmental in nature, placing emphasis on derangements occurring at the level of intra- and intercellular signaling during corticogenesis. At present, there is an ever-growing list of mutations in putative susceptibility genes in affected individuals, preventing effective transformation of knowledge gathered from basic science research to the clinic. In response, the focus of ASD biology has shifted toward the identification of cellular signaling pathways that are common to various ASD-related mutations in hopes that these shared pathways may serve as more promising treatment targets than targeting individual genes or proteins. To this end, the endogenous cannabinoid (endocannabinoid, eCB) system has recently emerged as a promising therapeutic target in the field of ASD research. The eCB system is altered in several neurological disorders, but the role of these bioactive lipids in ASD etiology remains poorly understood. In this perspective, we review current evidence linking eCB signaling to ASDs and put forth the notion that continued focus on eCBs in autism research may provide valuable insight into pathophysiology and treatment strategies. In addition to its role in modulating transmitter release at mature synapses, the eCB signaling system plays important roles in many aspects of cortical development, and disruption of these effects of eCBs may also be related to ASD pathophysiology.
Collapse
Affiliation(s)
- Mason L Yeh
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06032, USA
| | - Eric S Levine
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06032, USA
| |
Collapse
|