1
|
Fatt MP, Zhang MD, Kupari J, Altınkök M, Yang Y, Hu Y, Svenningsson P, Ernfors P. Morphine-responsive neurons that regulate mechanical antinociception. Science 2024; 385:eado6593. [PMID: 39208104 PMCID: PMC7616448 DOI: 10.1126/science.ado6593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 07/03/2024] [Indexed: 09/04/2024]
Abstract
Opioids are widely used, effective analgesics to manage severe acute and chronic pain, although they have recently come under scrutiny because of epidemic levels of abuse. While these compounds act on numerous central and peripheral pain pathways, the neuroanatomical substrate for opioid analgesia is not fully understood. By means of single-cell transcriptomics and manipulation of morphine-responsive neurons, we have identified an ensemble of neurons in the rostral ventromedial medulla (RVM) that regulates mechanical nociception in mice. Among these, forced activation or silencing of excitatory RVMBDNF projection neurons mimicked or completely reversed morphine-induced mechanical antinociception, respectively, via a brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB)-dependent mechanism and activation of inhibitory spinal galanin-positive neurons. Our results reveal a specific RVM-spinal circuit that scales mechanical nociception whose function confers the antinociceptive properties of morphine.
Collapse
Affiliation(s)
- Michael P. Fatt
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65Stockholm, Sweden
| | - Ming-Dong Zhang
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65Stockholm, Sweden
| | - Jussi Kupari
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65Stockholm, Sweden
| | - Müge Altınkök
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65Stockholm, Sweden
| | - Yunting Yang
- Division of Neuro, Department of Clinical Neuroscience, Karolinska Institutet, 171 77Stockholm, Sweden
| | - Yizhou Hu
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65Stockholm, Sweden
| | - Per Svenningsson
- Division of Neuro, Department of Clinical Neuroscience, Karolinska Institutet, 171 77Stockholm, Sweden
| | - Patrik Ernfors
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65Stockholm, Sweden
| |
Collapse
|
2
|
Zhu ZA, Li YY, Xu J, Xue H, Feng X, Zhu YC, Xiong ZQ. CDKL5 deficiency in adult glutamatergic neurons alters synaptic activity and causes spontaneous seizures via TrkB signaling. Cell Rep 2023; 42:113202. [PMID: 37777961 DOI: 10.1016/j.celrep.2023.113202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/26/2023] [Accepted: 09/16/2023] [Indexed: 10/03/2023] Open
Abstract
CDKL5 deficiency disorder (CDD) is a severe epileptic encephalopathy resulting from pathological mutations in the X-linked cyclin-dependent kinase-like 5 (CDKL5) gene. Despite significant progress in understanding the neuronal function of CDKL5, the molecular mechanisms underlying CDD-associated epileptogenesis are unknown. Here, we report that acute ablation of CDKL5 from adult forebrain glutamatergic neurons leads to elevated neural network activity in the dentate gyrus and the occurrence of early-onset spontaneous seizures via tropomyosin-related kinase B (TrkB) signaling. We observe increased expression of brain-derived neurotrophic factor (BDNF) and enhanced activation of its receptor TrkB in the hippocampus of Cdkl5-deficient mice prior to the onset of behavioral seizures. Moreover, reducing TrkB signaling in these mice rescues the altered synaptic activity and suppresses recurrent seizures. These results suggest that TrkB signaling mediates epileptogenesis in a mouse model of CDD and that targeting this pathway might be effective for treating epilepsy in patients affected by CDKL5 mutations.
Collapse
Affiliation(s)
- Zi-Ai Zhu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi-Yan Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Juan Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai 200031, China
| | - Hui Xue
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xue Feng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai 200031, China
| | - Yong-Chuan Zhu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| | - Zhi-Qi Xiong
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
3
|
Egbenya DL, Hussain S, Lai YC, Anderson AE, Davanger S. Synapse-specific changes in Arc and BDNF in rat hippocampus following chronic temporal lobe epilepsy. Neurosci Res 2022; 191:1-12. [PMID: 36535366 DOI: 10.1016/j.neures.2022.12.006] [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: 06/22/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Expression of immediate early genes (IEGs) in the brain is important for synaptic plasticity, and probably also in neurodegenerative conditions. To understand the cellular mechanisms of the underlying neuropathophysiological processes in epilepsy, we need to pinpoint changes in concentration of synaptic plasticity-related proteins at subsynaptic levels. In this study, we examined changes in synaptic expression of Activity-regulated cytoskeleton-associated (Arc) and Brai Derived Neurotrophic Factor (BDNF) in a rat model of kainate-induced temporal lobe epilepsy (TLE). Western blotting showed reduced concentrations of Arc and increased concentrations of BDNF in hippocampal synaptosomes in chronic TLE rats. Then, using quantitative electron microscopy, we found corresponding changes in subsynaptic regions in the hippocampus. Specifically, we detected significant reductions in the concentrations of Arc in the presynaptic terminal of Schaffer collateral glutamatergic synapses in the stratum radiatum of the CA1 area in TLE, as well as in their adjacent postsynaptic spines. In CA3, there was a significant reduction of Arc only in the presynaptic terminal cytoplasm. Conversely, in CA3, there was a significant increase in the expression of BDNF in the presynaptic terminal, but not in the postsynaptic spine. Significant increase in BDNF concentration in the CA1 postsynaptic density was also obtained. We hypothesize that the observed changes in Arc and BDNF may contribute to both cognitive impairment and increased excitotoxic vulnerability in chronic epilepsy.
Collapse
Affiliation(s)
- Daniel L Egbenya
- Laboratory for Synaptic Plasticity, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Physiology, School of Medical Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Suleman Hussain
- Laboratory for Synaptic Plasticity, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway; Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway.
| | - Yi-Chen Lai
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Anne E Anderson
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Svend Davanger
- Laboratory for Synaptic Plasticity, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
4
|
Mehterov N, Minchev D, Gevezova M, Sarafian V, Maes M. Interactions Among Brain-Derived Neurotrophic Factor and Neuroimmune Pathways Are Key Components of the Major Psychiatric Disorders. Mol Neurobiol 2022; 59:4926-4952. [PMID: 35657457 DOI: 10.1007/s12035-022-02889-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/17/2022] [Indexed: 10/25/2022]
Abstract
The purpose of this review is to summarize the current knowledge regarding the reciprocal associations between brain-derived neurotrophic factor (BDNF) and immune-inflammatory pathways and how these links may explain the involvement of this neurotrophin in the immune pathophysiology of mood disorders and schizophrenia. Toward this end, we delineated the protein-protein interaction (PPI) network centered around BDNF and searched PubMed, Scopus, Google Scholar, and Science Direct for papers dealing with the involvement of BDNF in the major psychosis, neurodevelopment, neuronal functions, and immune-inflammatory and related pathways. The PPI network was built based on the significant interactions of BDNF with neurotrophic (NTRK2, NTF4, and NGFR), immune (cytokines, STAT3, TRAF6), and cell-cell junction (CTNNB, CDH1) DEPs (differentially expressed proteins). Enrichment analysis shows that the most significant terms associated with this PPI network are the tyrosine kinase receptor (TRKR) and Src homology region two domain-containing phosphatase-2 (SHP2) pathways, tyrosine kinase receptor signaling pathways, positive regulation of kinase and transferase activity, cytokine signaling, and negative regulation of the immune response. The participation of BDNF in the immune response and its interactions with neuroprotective and cell-cell adhesion DEPs is probably a conserved regulatory process which protects against the many detrimental effects of immune activation and hyperinflammation including neurotoxicity. Lowered BDNF levels in mood disorders and schizophrenia (a) are associated with disruptions in neurotrophic signaling and activated immune-inflammatory pathways leading to neurotoxicity and (b) may interact with the reduced expression of other DEPs (CTNNB1, CDH1, or DISC1) leading to multiple aberrations in synapse and axonal functions.
Collapse
Affiliation(s)
- Nikolay Mehterov
- Department of Medical Biology, Medical University of Plovdiv, Plovdiv, Bulgaria.,Research Institute at Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Danail Minchev
- Department of Medical Biology, Medical University of Plovdiv, Plovdiv, Bulgaria.,Research Institute at Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Maria Gevezova
- Department of Medical Biology, Medical University of Plovdiv, Plovdiv, Bulgaria.,Research Institute at Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Victoria Sarafian
- Department of Medical Biology, Medical University of Plovdiv, Plovdiv, Bulgaria.,Research Institute at Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Michael Maes
- Faculty of Medicine, Department of Psychiatry, Chulalongkorn University, Bangkok, 10330, Thailand. .,Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria. .,Department of Psychiatry, IMPACT Strategic Research Centre, Deakin University, Geelong, VIC, Australia.
| |
Collapse
|
5
|
Birdsall V, Kirwan K, Zhu M, Imoto Y, Wilson SM, Watanabe S, Waites CL. Axonal transport of Hrs is activity dependent and facilitates synaptic vesicle protein degradation. Life Sci Alliance 2022; 5:5/10/e202000745. [PMID: 35636965 PMCID: PMC9152131 DOI: 10.26508/lsa.202000745] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 11/29/2022] Open
Abstract
This study describes an activity-dependent mechanism for transporting ESCRT-0 protein Hrs to synaptic vesicle (SV) pools, facilitating SV protein degradation in response to increased neuronal firing. Turnover of synaptic vesicle (SV) proteins is vital for the maintenance of healthy and functional synapses. SV protein turnover is driven by neuronal activity in an endosomal sorting complex required for transport (ESCRT)-dependent manner. Here, we characterize a critical step in this process: axonal transport of ESCRT-0 component Hrs, necessary for sorting proteins into the ESCRT pathway and recruiting downstream ESCRT machinery to catalyze multivesicular body (MVB) formation. We find that neuronal activity stimulates the formation of presynaptic endosomes and MVBs, as well as the motility of Hrs+ vesicles in axons and their delivery to SV pools. Hrs+ vesicles co-transport ESCRT-0 component STAM1 and comprise a subset of Rab5+ vesicles, likely representing pro-degradative early endosomes. Furthermore, we identify kinesin motor protein KIF13A as essential for the activity-dependent transport of Hrs to SV pools and the degradation of SV membrane proteins. Together, these data demonstrate a novel activity- and KIF13A-dependent mechanism for mobilizing axonal transport of ESCRT machinery to facilitate the degradation of SV membrane proteins.
Collapse
Affiliation(s)
- Veronica Birdsall
- Neurobiology and Behavior PhD Program, Columbia University, New York, NY, USA
| | - Konner Kirwan
- Neurobiology and Behavior PhD Program, Columbia University, New York, NY, USA
| | - Mei Zhu
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Yuuta Imoto
- Department of Cell Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Scott M Wilson
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shigeki Watanabe
- Department of Cell Biology, Johns Hopkins University, Baltimore, MD, USA.,Solomon H Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Clarissa L Waites
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA .,Department of Neuroscience, Columbia University, New York, NY, USA
| |
Collapse
|
6
|
Neurotrophin Signaling Impairment by Viral Infections in the Central Nervous System. Int J Mol Sci 2022; 23:ijms23105817. [PMID: 35628626 PMCID: PMC9146244 DOI: 10.3390/ijms23105817] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 02/04/2023] Open
Abstract
Neurotrophins, such as nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and neurotrophin 3 (NT-3), NT-4, and NT-5, are proteins involved in several important functions of the central nervous system. The activation of the signaling pathways of these neurotrophins, or even by their immature form, pro-neurotrophins, starts with their recognition by cellular receptors, such as tropomyosin receptor kinase (Trk) and 75 kD NT receptors (p75NTR). The Trk receptor is considered to have a high affinity for attachment to specific neurotrophins, while the p75NTR receptor has less affinity for attachment with neurotrophins. The correct functioning of these signaling pathways contributes to proper brain development, neuronal survival, and synaptic plasticity. Unbalanced levels of neurotrophins and pro-neurotrophins have been associated with neurological disorders, illustrating the importance of these molecules in the central nervous system. Furthermore, reports have indicated that viruses can alter the normal levels of neurotrophins by interfering with their signaling pathways. This work discusses the importance of neurotrophins in the central nervous system, their signaling pathways, and how viruses can affect them.
Collapse
|
7
|
Stojanovic T, Velarde Gamez D, Schuld GJ, Bormann D, Cabatic M, Uhrin P, Lubec G, Monje FJ. Age-Dependent and Pathway-Specific Bimodal Action of Nicotine on Synaptic Plasticity in the Hippocampus of Mice Lacking the miR-132/212 Genes. Cells 2022; 11:261. [PMID: 35053378 PMCID: PMC8774101 DOI: 10.3390/cells11020261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 01/06/2022] [Accepted: 01/11/2022] [Indexed: 12/19/2022] Open
Abstract
Nicotine addiction develops predominantly during human adolescence through smoking. Self-administration experiments in rodents verify this biological preponderance to adolescence, suggesting evolutionary-conserved and age-defined mechanisms which influence the susceptibility to nicotine addiction. The hippocampus, a brain region linked to drug-related memory storage, undergoes major morpho-functional restructuring during adolescence and is strongly affected by nicotine stimulation. However, the signaling mechanisms shaping the effects of nicotine in young vs. adult brains remain unclear. MicroRNAs (miRNAs) emerged recently as modulators of brain neuroplasticity, learning and memory, and addiction. Nevertheless, the age-dependent interplay between miRNAs regulation and hippocampal nicotinergic signaling remains poorly explored. We here combined biophysical and pharmacological methods to examine the impact of miRNA-132/212 gene-deletion (miRNA-132/212-/-) and nicotine stimulation on synaptic functions in adolescent and mature adult mice at two hippocampal synaptic circuits: the medial perforant pathway (MPP) to dentate yrus (DG) synapses (MPP-DG) and CA3 Schaffer collaterals to CA1 synapses (CA3-CA1). Basal synaptic transmission and short-term (paired-pulse-induced) synaptic plasticity was unaltered in adolescent and adult miRNA-132/212-/- mice hippocampi, compared with wild-type controls. However, nicotine stimulation promoted CA3-CA1 synaptic potentiation in mature adult (not adolescent) wild-type and suppressed MPP-DG synaptic potentiation in miRNA-132/212-/- mice. Altered levels of CREB, Phospho-CREB, and acetylcholinesterase (AChE) expression were further detected in adult miRNA-132/212-/- mice hippocampi. These observations propose miRNAs as age-sensitive bimodal regulators of hippocampal nicotinergic signaling and, given the relevance of the hippocampus for drug-related memory storage, encourage further research on the influence of miRNAs 132 and 212 in nicotine addiction in the young and the adult brain.
Collapse
Affiliation(s)
- Tamara Stojanovic
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria; (D.V.G.); (G.J.S.); (D.B.); (M.C.)
| | - David Velarde Gamez
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria; (D.V.G.); (G.J.S.); (D.B.); (M.C.)
| | - Gabor Jorrid Schuld
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria; (D.V.G.); (G.J.S.); (D.B.); (M.C.)
| | - Daniel Bormann
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria; (D.V.G.); (G.J.S.); (D.B.); (M.C.)
- Laboratory for Cardiac and Thoracic Diagnosis, Department of Surgery, Regeneration and Applied Immunology, Medical University of Vienna, Research Laboratories Vienna General Hospital, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Division of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Maureen Cabatic
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria; (D.V.G.); (G.J.S.); (D.B.); (M.C.)
| | - Pavel Uhrin
- Center for Physiology and Pharmacology, Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, 1090 Vienna, Austria;
| | - Gert Lubec
- Department of Neuroproteomics, Paracelsus Medical University, 5020 Salzburg, Austria;
| | - Francisco J. Monje
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria; (D.V.G.); (G.J.S.); (D.B.); (M.C.)
| |
Collapse
|
8
|
Golitabari N, Mohammadian F, Salari AA, Amani M. Neonatal NMDA blockade alters the LTP, LTD and cognitive functions in male and female Wistar rats. Neuropharmacology 2021; 205:108896. [PMID: 34822815 DOI: 10.1016/j.neuropharm.2021.108896] [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: 09/14/2021] [Revised: 10/31/2021] [Accepted: 11/19/2021] [Indexed: 12/31/2022]
Abstract
There is compelling evidence that neonatal blockade of NMDA receptors by phencyclidine (PCP) is associated with cognitive impairment in adulthood but little is known about the effects of early life PCP treatment on synaptic function later in life. Here, we sought to determine whether early life exposure to PCP alters the electrophysiologic function of hippocampal CA1 neurons in adult rats. To this end, male and female Wistar rats received either saline or PCP (10 mg/kg) on postnatal days (PND) 7, 9, and 11, and then underwent separate behavioral and electrophysiology tests in adulthood. Neonatal PCP treatment did not alter basic synaptic transmission and had only a modest effect on frequency following (FF) capacity but significantly decreased the paired-pulse facilitation (PPF) in the Schaffer collateral (SC)-CA1 pathway. We found that PCP treatment significantly attenuated the long-term potentiation (LTP) and long-term depression (LTD) in CA1 neurons accompanied by pronounced alteration in complex response profile in adult rats. The electrophysiology data were comparable in male and female rats and reliably associated with impaired spatial reference and working memories in these animals. Overall, this study suggests that blockade of NMDA receptors during early life deteriorates the short-term and long-term synaptic plasticity and complex response profile of CA1 neurons in adulthood.
Collapse
Affiliation(s)
- Nastaran Golitabari
- Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Forouzan Mohammadian
- Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali-Akbar Salari
- Salari Institute of Cognitive and Behavioral Disorders (SICBD), Karaj, Alborz, Iran
| | - Mohammad Amani
- Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
9
|
Thorsdottir D, Einwag Z, Erdos B. BDNF shifts excitatory-inhibitory balance in the paraventricular nucleus of the hypothalamus to elevate blood pressure. J Neurophysiol 2021; 126:1209-1220. [PMID: 34406887 DOI: 10.1152/jn.00247.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Presympathetic neurons in the paraventricular nucleus of the hypothalamus (PVN) play a key role in cardiovascular regulation. We have previously shown that brain-derived neurotrophic factor (BDNF), acting in the PVN, increases sympathetic activity and blood pressure and serves as a key regulator of stress-induced hypertensive responses. BDNF is known to alter glutamatergic and GABA-ergic signaling broadly in the central nervous system, but whether BDNF has similar actions in the PVN remains to be investigated. Here, we tested the hypothesis that increased BDNF expression in the PVN elevates blood pressure by enhancing N-methyl-d-aspartate (NMDA) receptor (NMDAR)- and inhibiting GABAA receptor (GABAAR)-mediated signaling. Sprague-Dawley rats received bilateral PVN injections of AAV2 viral vectors expressing green fluorescent protein (GFP) or BDNF. Three weeks later, cardiovascular responses to PVN injections of NMDAR and GABAAR agonists and antagonists were recorded under α-chloralose-urethane anesthesia. In addition, expressions of excitatory and inhibitory signaling components in the PVN were assessed using immunofluorescence. Our results showed that NMDAR inhibition led to a greater decrease in blood pressure in the BDNF vs. GFP group, while GABAAR inhibition led to greater increases in blood pressure in the GFP group compared to BDNF. Conversely, GABAAR activation decreased blood pressure significantly more in GFP vs. BDNF rats. In addition, immunoreactivity of NMDAR1 was upregulated, while GABAAR-α1 and K+/Cl- cotransporter 2 were downregulated by BDNF overexpression in the PVN. In summary, our findings indicate that hypertensive actions of BDNF within the PVN are mediated, at least in part, by augmented NMDAR and reduced GABAAR signaling.NEW & NOTEWORTHY We have shown that BDNF, acting in the PVN, elevates blood pressure in part by augmenting NMDA receptor-mediated excitatory input and by diminishing GABAA receptor-mediated inhibitory input to PVN neurons. In addition, we demonstrate that elevated BDNF expression in the PVN upregulates NMDA receptor immunoreactivity and downregulates GABAA receptor as well as KCC2 transporter immunoreactivity.
Collapse
Affiliation(s)
| | - Zachary Einwag
- Department of Pharmacology, University of Vermont, Burlington, Vermont
| | - Benedek Erdos
- Department of Pharmacology, University of Vermont, Burlington, Vermont
| |
Collapse
|
10
|
Sieck GC, Gransee HM, Zhan WZ, Mantilla CB. Acute intrathecal BDNF enhances functional recovery after cervical spinal cord injury in rats. J Neurophysiol 2021; 125:2158-2165. [PMID: 33949892 DOI: 10.1152/jn.00146.2021] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Unilateral C2 hemisection (C2SH) disrupts descending inspiratory-related drive to phrenic motor neurons and thus, silences rhythmic diaphragm muscle (DIAm) activity. There is gradual recovery of rhythmic DIAm EMG activity over time post-C2SH, consistent with neuroplasticity, which is enhanced by chronic (2 wk) intrathecal BDNF treatment. In the present study, we hypothesized that acute (30 min) intrathecal BDNF treatment also enhances recovery of DIAm EMG activity after C2SH. Rats were implanted with bilateral DIAm EMG electrodes to verify the absence of ipsilateral eupneic DIAm EMG activity at the time of C2SH and at 3 days post-C2SH. In those animals displaying no recovery of DIAm EMG activity after 28 days (n = 7), BDNF was administered intrathecally (450 mcg) at C4. DIAm EMG activity was measured continuously both before and for 30 min after BDNF treatment, during eupnea, hypoxia-hypercapnia, and spontaneous sighs. Acute BDNF treatment restored eupneic DIAm EMG activity in all treated animals to an amplitude that was 78% ± 9% of pre-C2SH root mean square (RMS) (P < 0.001). In addition, acute BDNF treatment increased DIAm RMS EMG amplitude during hypoxia-hypercapnia (P = 0.023) but had no effect on RMS EMG amplitude during sighs. These results support an acute modulatory role of BDNF signaling on excitatory synaptic transmission at phrenic motor neurons after cervical spinal cord injury.NEW & NOTEWORTHY Brain-derived neurotrophic factor (BDNF) plays an important role in promoting neuroplasticity following unilateral C2 spinal hemisection (C2SH). BDNF was administered intrathecally in rats displaying lack of ipsilateral inspiratory-related diaphragm (DIAm) EMG activity after C2SH. Acute BDNF treatment (30 min) restored eupneic DIAm EMG activity in all treated animals to 78% ± 9% of pre-C2SH level. In addition, acute BDNF treatment increased DIAm EMG amplitude during hypoxia-hypercapnia but had no effect on EMG amplitude during sighs.
Collapse
Affiliation(s)
- Gary C Sieck
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota.,Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Heather M Gransee
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Wen-Zhi Zhan
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Carlos B Mantilla
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota.,Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
11
|
Martínez-Torres NI, Vázquez-Hernández N, Martín-Amaya-Barajas FL, Flores-Soto M, González-Burgos I. Ibotenic acid induced lesions impair the modulation of dendritic spine plasticity in the prefrontal cortex and amygdala, a phenomenon that underlies working memory and social behavior. Eur J Pharmacol 2021; 896:173883. [PMID: 33513334 DOI: 10.1016/j.ejphar.2021.173883] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 01/08/2023]
Abstract
The lesions induced by Ibotenic acid (IA) emulate some of the symptoms associated with schizophrenia, such as impaired working memory that is predominantly organized by the medial prefrontal cortex (mPFC), or difficulties in social interactions that aremainly organized by the amygdala (AMG). The plastic capacity of dendritic spines in neurons of the mPFC and AMG is modulated by molecules that participate in the known deterioration of working memory, although the influence of these on the socialization of schizophrenic patients is unknown. Here, the effect of a neonatal IA induced lesion on social behavior and working memory was evaluated in adult rats, along with the changes in cytoarchitecture of dendritic spines and their protein content, specifically the postsynaptic density protein 95 (PSD-95), Synaptophysin (Syn), AMPA receptors, and brain-derived neurotrophic factor (BDNF). Both working memory and social behavior were impaired, and the density of the spines, as well as their PSD-95, Syn, AMPA receptor and BDNF content was lower in IA lesioned animals. The proportional density of thin, mushroom, stubby and wide spines resulted in plastic changes that suggest the activation of compensatory processes in the face of the adverse effects of the lesion. In addition, the reduction in the levels of the modulating factors also suggests that the signaling pathways in which such factors are implicated would be altered in the brains of patients with schizophrenia. Accordingly, the experimental study of such signaling pathways is likely to aid the development of more effective pharmacological strategies for the treatment of schizophrenia.
Collapse
Affiliation(s)
- Néstor I Martínez-Torres
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Guadalajara, Jal., Mexico; Centro Universitario del Norte, Universidad de Guadalajara, Colotlán, Jal., Mexico
| | - Nallely Vázquez-Hernández
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Guadalajara, Jal., Mexico
| | | | - Mario Flores-Soto
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Guadalajara, Jal., Mexico
| | - Ignacio González-Burgos
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Guadalajara, Jal., Mexico.
| |
Collapse
|
12
|
Flores-Soto M, Romero-Guerrero C, Vázquez-Hernández N, Tejeda-Martínez A, Martín-Amaya-Barajas FL, Orozco-Suárez S, González-Burgos I. Pentylenetetrazol-induced seizures in adult rats are associated with plastic changes to the dendritic spines on hippocampal CA1 pyramidal neurons. Behav Brain Res 2021; 406:113198. [PMID: 33657439 DOI: 10.1016/j.bbr.2021.113198] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/29/2022]
Abstract
Epilepsy is a chronic neurobehavioral disorder whereby an imbalance between neurochemical excitation and inhibition at the synaptic level provokes seizures. Various experimental models have been used to study epilepsy, including that based on acute or chronic administration of Pentylenetetrazol (PTZ). In this study, a single PTZ dose (60 mg/kg) was administered to adult male rats and 30 min later, various neurobiological parameters were studied related to the transmission and modulation of excitatory impulses in pyramidal neurons of the hippocampal CA1 field. Rats experienced generalized seizures 1-3 min after PTZ administration, accompanied by elevated levels of Synaptophysin and Glutaminase. This response suggests presynaptic glutamate release is exacerbated to toxic levels, which eventually provokes neuronal death as witnessed by the higher levels of Caspase-3, TUNEL and GFAP. Similarly, the increase in PSD-95 suggests that viable dendritic spines are functional. Indeed, the increase in stubby and wide spines is likely related to de novo spinogenesis, and the regulation of neuronal excitability, which could represent a plastic response to the synaptic over-excitation. Furthermore, the increase in mushroom spines could be associated with the storage of cognitive information and the potentiation of thin spines until they are transformed into mushroom spines. However, the reduction in BDNF suggests that the activity of these spines would be down-regulated, may in part be responsible for the cognitive decline related to hippocampal function in patients with epilepsy.
Collapse
Affiliation(s)
- Mario Flores-Soto
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara, Jal., Mexico
| | - Christian Romero-Guerrero
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara, Jal., Mexico
| | - Nallely Vázquez-Hernández
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara, Jal., Mexico
| | - Aldo Tejeda-Martínez
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara, Jal., Mexico
| | | | - Sandra Orozco-Suárez
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades, CMN S-XXI, IMSS, Guadalajara, Jal., Mexico
| | - Ignacio González-Burgos
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara, Jal., Mexico.
| |
Collapse
|
13
|
Xue Y, Liang H, Yang R, Deng K, Tang M, Zhang M. The role of pro- and mature neurotrophins in the depression. Behav Brain Res 2021; 404:113162. [PMID: 33549684 DOI: 10.1016/j.bbr.2021.113162] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 01/22/2021] [Accepted: 02/02/2021] [Indexed: 12/24/2022]
Abstract
Neurotrophic factors, which can provide nutritional support to neurons and neuronal cells, also played an important role in their proliferation and survival. As signaling molecules, it also mediated the learning, memory and other activities in the brain. The latest study shows that neurotrophic factors have diametrically opposing effects of the pro- and mature form through distinct receptors. In this review, we summarize the different forms of neurotrophic factors, related receptors, and the corresponding biological effects. More importantly, we expounded the physiology and pathology mechanisms of brain-derived neurotrophic factor(BDNF)in depression. It is hopefully to provide new idea on the relationship of neurotrophic factors and depression.
Collapse
Affiliation(s)
- Ying Xue
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Hongyan Liang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Rui Yang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Kunhong Deng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.
| | - Mimi Tang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Mengqi Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
14
|
Limited Sensitivity of Hippocampal Synaptic Function or Network Oscillations to Unmodulated Kilohertz Electric Fields. eNeuro 2020; 7:ENEURO.0368-20.2020. [PMID: 33328248 PMCID: PMC7773889 DOI: 10.1523/eneuro.0368-20.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 11/21/2022] Open
Abstract
Understanding the cellular mechanisms of kilohertz (kHz) electrical stimulation is of broad interest in neuromodulation including forms of transcranial electrical stimulation, interferential stimulation, and high-rate spinal cord stimulation (SCS). Yet, the well-established low-pass filtering by neuronal membranes suggests minimal neuronal polarization in respond to charge-balanced kHz stimulation. The hippocampal brain slice model is among the most studied systems in neuroscience and exhaustively characterized in screening the effects of electrical stimulation. High-frequency electric fields of varied amplitudes (1–150 V/m), waveforms (sinusoidal, symmetrical pule, asymmetrical pulse) and frequencies (1 and10 kHz) were tested. Changes in single or paired-pulse field EPSPs (fEPSP) in CA1 were measured in response to radial-directed and tangential-directed electric fields, with brief (30 s) or long (30 min) application times. The effects of kHz stimulation on ongoing endogenous network activity were tested in carbachol-induced γ oscillation of CA3a and CA3c. Across 23 conditions evaluated, no significant changes in fEPSP were resolved, while responses were detected for within-slice control direct current (DC) fields; 1-kHz sinusoidal and pulse stimulation (≥60 V/m), but not 10 kHz, induced changes in oscillating neuronal network. We thus report no responses to low-amplitude 1-kHz or any 10-kHz fields, suggesting that any brain sensitivity to these fields is via yet to be-determined mechanism(s) of action which were not identified in our experimental preparation.
Collapse
|
15
|
Obesity is Associated with Reduced Plasticity of the Human Motor Cortex. Brain Sci 2020; 10:brainsci10090579. [PMID: 32839377 PMCID: PMC7564681 DOI: 10.3390/brainsci10090579] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/13/2020] [Accepted: 08/17/2020] [Indexed: 01/06/2023] Open
Abstract
Obesity is characterised by excessive body fat and is associated with several detrimental health conditions, including cardiovascular disease and diabetes. There is some evidence that people who are obese have structural and functional brain alterations and cognitive deficits. It may be that these neurophysiological and behavioural consequences are underpinned by altered plasticity. This study investigated the relationship between obesity and plasticity of the motor cortex in people who were considered obese (n = 14, nine males, aged 35.4 ± 14.3 years) or healthy weight (n = 16, seven males, aged 26.3 ± 8.5 years). A brain stimulation protocol known as continuous theta burst transcranial magnetic stimulation was applied to the motor cortex to induce a brief suppression of cortical excitability. The suppression of cortical excitability was quantified using single-pulse transcranial magnetic stimulation to record and measure the amplitude of the motor evoked potential in a peripheral hand muscle. Therefore, the magnitude of suppression of the motor evoked potential by continuous theta burst stimulation was used as a measure of the capacity for plasticity of the motor cortex. Our results demonstrate that the healthy-weight group had a significant suppression of cortical excitability following continuous theta burst stimulation (cTBS), but there was no change in excitability for the obese group. Comparing the response to cTBS between groups demonstrated that there was an impaired plasticity response for the obese group when compared to the healthy-weight group. This might suggest that the capacity for plasticity is reduced in people who are obese. Given the importance of plasticity for human behaviour, our results add further emphasis to the potentially detrimental health effects of obesity.
Collapse
|
16
|
Hernandez-Echeagaray E. The role of the TrkB-T1 receptor in the neurotrophin-4/5 antagonism of brain-derived neurotrophic factor on corticostriatal synaptic transmission. Neural Regen Res 2020; 15:1973-1976. [PMID: 32394943 PMCID: PMC7716028 DOI: 10.4103/1673-5374.282224] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
This manuscript reviews the function and fundamental characteristics of the neurotrophins and their receptors to introduce the reader to the differential effects exhibited by the neurotrophins; brain-derived neurotrophic factor and neurotrophin 4/5 when acted together after sequential presentation. The neurotrophin 4/5 exhibits an inhibitory action on the modulatory effect of brain-derived neurotrophic factor in corticostriatal synapses when they are administered sequentially (brain-derived neurotrophic factor to neurotrophin 4/5). This inhibitory effect has not been previously documented and is relevant for these neurotrophins as both of them stimulate the TrkB receptor. The additive effect of these neurotrophins is also discussed and occurs when neurotrophin 4/5 exposure is followed by brain-derived neurotrophic factor in a mouse model of striatal degeneration. Occlusive and additive effects of both neurotrophins are accompanied by changes in the expression of the TrkB receptor isoforms, specifically TrkB-T1 and TrkB-FL, as well as differences in phosphorylation levels of the TrkB receptor. The results of the experiments described raise several questions to inquire about the role that TrkB-T1 receptor plays in striatal physiology, as well as the functional relevance of the interaction of brain-derived neurotrophic factor and neurotrophin 4/5 in the brain and more specifically at the striatal circuits in normal as well as pathological conditions.
Collapse
|
17
|
BDNF impact on synaptic dynamics: extra or intracellular long-term release differently regulates cultured hippocampal synapses. Mol Brain 2020; 13:43. [PMID: 32183860 PMCID: PMC7079446 DOI: 10.1186/s13041-020-00582-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 03/09/2020] [Indexed: 01/21/2023] Open
Abstract
Brain Derived Neurotrophic Factor (BDNF) signalling contributes to the formation, maturation and plasticity of Central Nervous System (CNS) synapses. Acute exposure of cultured brain circuits to BDNF leads to up-regulation of glutamatergic neuro-transmission, by the accurate tuning of pre and post synaptic features, leading to structural and functional synaptic changes. Chronic BDNF treatment has been comparatively less investigated, besides it may represent a therapeutic option to obtain rescue of post-injury alterations of synaptic networks. In this study, we used a paradigm of BDNF long-term (4 days) incubation to assess in hippocampal neurons in culture, the ability of such a treatment to alter synapses. By patch clamp recordings we describe the augmented function of excitatory neurotransmission and we further explore by live imaging the presynaptic changes brought about by long-term BDNF. In our study, exogenous long-term BDNF exposure of post-natal neurons did not affect inhibitory neurotransmission. We further compare, by genetic manipulations of cultured neurons and BDNF release, intracellular overexpression of this neurotrophin at the same developmental age. We describe for the first-time differences in synaptic modulation by BDNF with respect to exogenous or intracellular release paradigms. Such a finding holds the potential of influencing the design of future therapeutic strategies.
Collapse
|
18
|
Deyama S, Bang E, Kato T, Li XY, Duman RS. Neurotrophic and Antidepressant Actions of Brain-Derived Neurotrophic Factor Require Vascular Endothelial Growth Factor. Biol Psychiatry 2019; 86:143-152. [PMID: 30712809 PMCID: PMC6597338 DOI: 10.1016/j.biopsych.2018.12.014] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/17/2018] [Accepted: 12/17/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND Activity-dependent release of brain-derived neurotrophic factor (BDNF) in the medial prefrontal cortex (mPFC) is essential for the rapid and sustained antidepressant actions of ketamine, and a recent study shows a similar requirement for vascular endothelial growth factor (VEGF). Since BDNF is reported to stimulate VEGF expression and/or release in neuroblastoma cells, the present study tested the hypothesis that the actions of BDNF are mediated by VEGF. METHODS The role of VEGF in the antidepressant behavioral actions of BDNF was tested by intra-mPFC coinfusion of a VEGF neutralizing antibody and by neuron-specific deletion of VEGF. The influence of BDNF on the release of VEGF and the role of VEGF in the neurotrophic actions of BDNF were determined in rat primary cortical neurons. The role of BDNF in the behavioral and neurotrophic actions of VEGF was also determined. RESULTS The results show that the rapid and sustained antidepressant-like actions of intra-mPFC BDNF are blocked by coinfusion of a VEGF neutralizing antibody, and that neuron-specific mPFC deletion of VEGF blocks the antidepressant-like actions of BDNF. Studies in primary cortical neurons demonstrate that BDNF stimulates the release of VEGF and that BDNF induction of dendrite complexity is blocked by a selective VEGF-fetal liver kinase 1 receptor antagonist. Surprisingly, the results also show reciprocal interactions, indicating that the behavioral and neurotrophic actions of VEGF are dependent on BDNF. CONCLUSIONS These findings indicate that the antidepressant-like and neurotrophic actions of BDNF require VEGF signaling, but they also demonstrate reciprocal interdependence for BDNF in the actions of VEGF.
Collapse
Affiliation(s)
- Satoshi Deyama
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA.,Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa 920-1192, Japan
| | - Eunyoung Bang
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Taro Kato
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA.,Drug Development Research Laboratories, Sumitomo Dainippon Pharma Co., Ltd., Suita 564-0053, Japan
| | - Xiao-Yuan Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Ronald S. Duman
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA.,Correspondence: Ronald S. Duman, Ph.D., Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT 06519, USA. Tel: 203-974-7726, Fax: 203-974-7724,
| |
Collapse
|
19
|
Cuzon Carlson VC, Ford MM, Carlson TL, Lomniczi A, Grant KA, Ferguson B, Cervera-Juanes RP. Modulation of Gpr39, a G-protein coupled receptor associated with alcohol use in non-human primates, curbs ethanol intake in mice. Neuropsychopharmacology 2019; 44:1103-1113. [PMID: 30610192 PMCID: PMC6461847 DOI: 10.1038/s41386-018-0308-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 12/13/2018] [Accepted: 12/26/2018] [Indexed: 12/17/2022]
Abstract
Alcohol use disorder (AUD) is a chronic condition with devastating health and socioeconomic effects. Still, pharmacotherapies to treat AUD are scarce. In a prior study aimed at identifying novel AUD therapeutic targets, we investigated the DNA methylome of the nucleus accumbens core (NAcc) of rhesus macaques after chronic alcohol use. The G-protein coupled receptor 39 (GPR39) gene was hypermethylated and its expression downregulated in heavy alcohol drinking macaques. GPR39 encodes a Zn2+-binding metabotropic receptor known to modulate excitatory and inhibitory neurotransmission, the balance of which is altered in AUD. These prior findings suggest that a GPR39 agonist would reduce alcohol intake. Using a drinking-in-the-dark two bottle choice (DID-2BC) model, we showed that an acute 7.5 mg/kg dose of the GPR39 agonist, TC-G 1008, reduced ethanol intake in mice without affecting total fluid intake, locomotor activity or saccharin preference. Furthermore, repeated doses of the agonist prevented ethanol escalation in an intermittent access 2BC paradigm (IA-2BC). This effect was reversible, as ethanol escalation followed agonist "wash out". As observed during the DID-2BC study, a subsequent acute agonist challenge during the IA-2BC procedure reduced ethanol intake by ~47%. Finally, Gpr39 activation was associated with changes in Gpr39 and Bdnf expression, and in glutamate release in the NAcc. Together, our findings suggest that GPR39 is a promising target for the development of prevention and treatment therapies for AUD.
Collapse
Affiliation(s)
- Verginia C Cuzon Carlson
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon, USA
| | - Matthew M Ford
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon, USA
| | - Timothy L Carlson
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, Oregon, USA
| | - Alejandro Lomniczi
- Division of Genetics, Oregon National Primate Research, Oregon Health and Sciences University, Beaverton, Oregon, USA
| | - Kathleen A Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon, USA
| | - Betsy Ferguson
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, Oregon, USA
- Division of Genetics, Oregon National Primate Research, Oregon Health and Sciences University, Beaverton, Oregon, USA
- Department of Molecular and Medical Genetics, Oregon Health and Sciences University, Portland, Oregon, USA
| | - Rita P Cervera-Juanes
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, Oregon, USA.
- Division of Genetics, Oregon National Primate Research, Oregon Health and Sciences University, Beaverton, Oregon, USA.
| |
Collapse
|
20
|
Prominent Postsynaptic and Dendritic Exocytosis of Endogenous BDNF Vesicles in BDNF-GFP Knock-in Mice. Mol Neurobiol 2019; 56:6833-6855. [DOI: 10.1007/s12035-019-1551-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/13/2019] [Indexed: 12/23/2022]
|
21
|
Torres-Cruz FM, César Vivar-Cortés I, Moran I, Mendoza E, Gómez-Pineda V, García-Sierra F, Hernández-Echeagaray E. NT-4/5 antagonizes the BDNF modulation of corticostriatal transmission: Role of the TrkB.T1 receptor. CNS Neurosci Ther 2019; 25:621-631. [PMID: 30666798 PMCID: PMC6488875 DOI: 10.1111/cns.13091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 11/14/2018] [Accepted: 11/19/2018] [Indexed: 12/14/2022] Open
Abstract
Neurotrophins are related to survival, growth, differentiation and neurotrophic maintenance as well as modulation of synaptic transmission in different regions of the nervous system. BDNF effects have been studied in the striatum due to the trophic role of BDNF in medium spiny neurons; however, less is known about the effects of NT‐4/5, which is also present in the striatum and activates the TrkB receptor along with BDNF. If both neurotrophins are present in the striatum, the following question arises: What role do they play in striatal physiology? Thus, the aim of this study was to determine the physiological effect of the sequential application and coexistence of BDNF and NT‐4/5 on the modulation of corticostriatal synapses. Our data demonstrated that neurotrophins exhibit differential effects depending on exposure order. BDNF did not modify NT‐4/5 effect; however, NT‐4/5 inhibited the effects of BDNF. Experiments carried out in COS‐7 cells to understand the mechanisms of this antagonism, indicated that NT‐4/5 exerts its inhibitory effect on BDNF by upregulating the TrkB.T1 and downregulating the TrkB‐FL isoforms of the TrkB receptor.
Collapse
Affiliation(s)
- Francisco M Torres-Cruz
- Laboratorio de Neurofisiología del Desarrollo y la Neurodegeneración, UBIMED, FES-Iztacala, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Israel César Vivar-Cortés
- Laboratorio de Neurofisiología del Desarrollo y la Neurodegeneración, UBIMED, FES-Iztacala, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Isaac Moran
- Laboratorio de Neurofisiología del Desarrollo y la Neurodegeneración, UBIMED, FES-Iztacala, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Ernesto Mendoza
- Laboratorio de Neurofisiología del Desarrollo y la Neurodegeneración, UBIMED, FES-Iztacala, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Victor Gómez-Pineda
- Laboratorio de Neurofisiología del Desarrollo y la Neurodegeneración, UBIMED, FES-Iztacala, Universidad Nacional Autónoma de México, Ciudad de México, México
| | | | - Elizabeth Hernández-Echeagaray
- Laboratorio de Neurofisiología del Desarrollo y la Neurodegeneración, UBIMED, FES-Iztacala, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
22
|
More JY, Bruna BA, Lobos PE, Galaz JL, Figueroa PL, Namias S, Sánchez GL, Barrientos GC, Valdés JL, Paula-Lima AC, Hidalgo C, Adasme T. Calcium Release Mediated by Redox-Sensitive RyR2 Channels Has a Central Role in Hippocampal Structural Plasticity and Spatial Memory. Antioxid Redox Signal 2018; 29:1125-1146. [PMID: 29357673 DOI: 10.1089/ars.2017.7277] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIMS Previous studies indicate that hippocampal synaptic plasticity and spatial memory processes entail calcium release from intracellular stores mediated by ryanodine receptor (RyR) channels. In particular, RyR-mediated Ca2+ release is central for the dendritic spine remodeling induced by brain-derived neurotrophic factor (BDNF), a neurotrophin that stimulates complex signaling pathways leading to memory-associated protein synthesis and structural plasticity. To examine if upregulation of ryanodine receptor type-2 (RyR2) channels and the spine remodeling induced by BDNF entail reactive oxygen species (ROS) generation, and to test if RyR2 downregulation affects BDNF-induced spine remodeling and spatial memory. RESULTS Downregulation of RyR2 expression (short hairpin RNA [shRNA]) in primary hippocampal neurons, or inhibition of nitric oxide synthase (NOS) or NADPH oxidase, prevented agonist-mediated RyR-mediated Ca2+ release, whereas BDNF promoted cytoplasmic ROS generation. RyR2 downregulation or inhibitors of N-methyl-d-aspartate (NMDA) receptors, or NOS or of NADPH oxidase type-2 (NOX2) prevented RyR2 upregulation and the spine remodeling induced by BDNF, as did incubation with the antioxidant agent N-acetyl l-cysteine. In addition, intrahippocampal injection of RyR2-directed antisense oligodeoxynucleotides, which caused significant RyR2 downregulation, caused conspicuous defects in a memorized spatial memory task. INNOVATION The present novel results emphasize the key role of redox-sensitive Ca2+ release mediated by RyR2 channels for hippocampal structural plasticity and spatial memory. CONCLUSION Based on these combined results, we propose (i) that BDNF-induced RyR2-mediated Ca2+ release and ROS generation via NOS/NOX2 are strictly required for the dendritic spine remodeling and the RyR2 upregulation induced by BDNF, and (ii) that RyR2 channel expression is crucial for spatial memory processes. Antioxid. Redox Signal. 29, 1125-1146.
Collapse
Affiliation(s)
- Jamileth Y More
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Barbara A Bruna
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Pedro E Lobos
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - José L Galaz
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Paula L Figueroa
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Silvia Namias
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Gina L Sánchez
- 2 Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Genaro C Barrientos
- 2 Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - José L Valdés
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile .,3 Department of Neuroscience, Faculty of Medicine, Universidad de Chile , Santiago, Chile
| | - Andrea C Paula-Lima
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile .,4 Institute for Research in Dental Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Cecilia Hidalgo
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile .,2 Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile .,3 Department of Neuroscience, Faculty of Medicine, Universidad de Chile , Santiago, Chile .,5 Center for Exercise , Metabolism and Cancer Studies, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Tatiana Adasme
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile .,6 Centro Integrativo de Biología y Química Aplicada, Universidad Bernardo O'Higgins , Santiago, Chile
| |
Collapse
|
23
|
Enhanced extinction of aversive memories in mice lacking SPARC-related protein containing immunoglobulin domains 1 (SPIG1/FSTL4). Neurobiol Learn Mem 2018; 152:61-70. [PMID: 29783061 DOI: 10.1016/j.nlm.2018.05.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/11/2018] [Accepted: 05/16/2018] [Indexed: 01/23/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) plays an important role in synaptic plasticity related to learning and memory. We previously reported that SPARC-related protein containing immunoglobulin domains 1 (SPIG1, also known as Follistatin-like protein 4, FSTL4) binds to pro-BDNF and negatively regulates BDNF maturation; however, its neurological functions, particularly in learning and memory, have not yet been elucidated. We herein examined the electrophysiological and behavioral phenotypes of Spig1-knockout (Spig1-KO) mice. Adult Spig1-KO mice exhibited greater excitability and facilitated long-term potentiation (LTP) in the CA1 region of hippocampal slices than age- and sex-matched wild-type (WT) mice. Facilitated LTP was reduced to the level of WT by the bath application of an anti-BDNF antibody to hippocampal slices. A step-through inhibitory avoidance learning paradigm revealed that the extinction of aversive memories was significantly enhanced in adult Spig1-KO mice, while they showed the normal acquisition of aversive memories; besides, spatial reference memory formation was also normal in the standard Morris water maze task. An intracerebroventricular (icv) injection of anti-BDNF in the process of extinction learning transiently induced the recurrence of aversive memories in Spig1-KO mice, but exerted no effects in WT mice. These results indicate a critical role for SPIG1 in BDNF-mediated synaptic plasticity in extinction of inhibitory avoidance memory.
Collapse
|
24
|
Brigadski T, Lichtenecker P, Lessmann V. Recording Activity-Dependent Release of BDNF from Hippocampal Neurons. BRAIN-DERIVED NEUROTROPHIC FACTOR (BDNF) 2018. [DOI: 10.1007/7657_2018_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
25
|
Kumar A, Pareek V, Faiq MA, Kumar P, Raza K, Prasoon P, Dantham S, Mochan S. Regulatory role of NGFs in neurocognitive functions. Rev Neurosci 2017; 28:649-673. [DOI: 10.1515/revneuro-2016-0031] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 01/25/2017] [Indexed: 12/16/2022]
Abstract
AbstractNerve growth factors (NGFs), especially the prototype NGF and brain-derived neurotrophic factor (BDNF), have a diverse array of functions in the central nervous system through their peculiar set of receptors and intricate signaling. They are implicated not only in the development of the nervous system but also in regulation of neurocognitive functions like learning, memory, synaptic transmission, and plasticity. Evidence even suggests their role in continued neurogenesis and experience-dependent neural network remodeling in adult brain. They have also been associated extensively with brain disorders characterized by neurocognitive dysfunction. In the present article, we aimed to make an exhaustive review of literature to get a comprehensive view on the role of NGFs in neurocognitive functions in health and disease. Starting with historical perspective, distribution in adult brain, implied molecular mechanisms, and developmental basis, this article further provides a detailed account of NGFs’ role in specified neurocognitive functions. Furthermore, it discusses plausible NGF-based homeostatic and adaptation mechanisms operating in the pathogenesis of neurocognitive disorders and has presents a survey of such disorders. Finally, it elaborates on current evidence and future possibilities in therapeutic applications of NGFs with an emphasis on recent research updates in drug delivery mechanisms. Conclusive remarks of the article make a strong case for plausible role of NGFs in comprehensive regulation of the neurocognitive functions and pathogenesis of related disorders and advocate that future research should be directed to explore use of NGF-based mechanisms in the prevention of implicated diseases as well as to target these molecules pharmacologically.
Collapse
Affiliation(s)
- Ashutosh Kumar
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
- Department of Anatomy, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Karaikal, Puducherry 609602, India
| | - Vikas Pareek
- Computational Neuroscience and Neuroimaging Division, National Brain Research Centre (NBRC), Manesar, Haryana 122051, India
| | - Muneeb A. Faiq
- Department of Ophthalmology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Pavan Kumar
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Khursheed Raza
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Pranav Prasoon
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Subrahamanyam Dantham
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Sankat Mochan
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| |
Collapse
|
26
|
Gulyaeva NV. Interplay between brain BDNF and glutamatergic systems: A brief state of the evidence and association with the pathogenesis of depression. BIOCHEMISTRY (MOSCOW) 2017; 82:301-307. [DOI: 10.1134/s0006297917030087] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
27
|
Yeh ML, Selvam R, Levine ES. BDNF-induced endocannabinoid release modulates neocortical glutamatergic neurotransmission. Synapse 2017; 71. [PMID: 28164368 DOI: 10.1002/syn.21962] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 01/23/2017] [Accepted: 02/01/2017] [Indexed: 01/07/2023]
Abstract
Endocannabinoids (eCBs) and neurotrophins, particularly brain-derived neurotrophic factor (BDNF), are potent neuromodulators found throughout the mammalian neocortex. Both eCBs and BDNF play critical roles in many behavioral and neurophysiological processes and are targets for the development of novel therapeutics. The effects of eCBs and BDNF are primarily mediated by the type 1 cannabinoid (CB1) receptor and the trkB tyrosine kinase receptor, respectively. Our laboratory and others have previously established that BDNF potentiates excitatory transmission by enhancing presynaptic glutamate release and modulating NMDA receptors. In contrast, we have shown that BDNF attenuates inhibitory transmission by inducing postsynaptic release of eCBs that act retrogradely to suppress GABA release in layer 2/3 of somatosensory cortex. Here, we hypothesized that BDNF also induces release of eCBs at excitatory synapses, which could have a mitigating or opposing effect on the direct presynaptic effects of BDNF. We found the highest levels of expression of CB1 and trkB and receptors in layers 2/3 and 5. Surprisingly, BDNF did not increase the frequency of spontaneous miniature excitatory postsynaptic currents (mEPSCs) onto layer 5 pyramidal neurons in somatosensory cortex, in contrast to its effects in the hippocampus and visual cortex. However, the effect of BDNF on mEPSC frequency in somatosensory cortex was unmasked by blocking CB1 receptors or disrupting eCB release. Thus, BDNF-trKB signaling regulates glutamate release in the somatosensory cortex via opposing effects, a direct presynaptic enhancement of release probability, and simultaneous postsynaptically-induced eCB release that decreases release probability via presynaptic CB1 receptors.
Collapse
Affiliation(s)
- Mason L Yeh
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT, 06030
| | - Rajamani Selvam
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT, 06030
| | - Eric S Levine
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT, 06030
| |
Collapse
|
28
|
Vedder LC, Savage LM. BDNF regains function in hippocampal long-term potentiation deficits caused by diencephalic damage. ACTA ACUST UNITED AC 2017; 24:81-85. [PMID: 28096497 PMCID: PMC5238722 DOI: 10.1101/lm.043927.116] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 11/02/2016] [Indexed: 01/13/2023]
Abstract
Thiamine deficiency (TD), commonly associated with chronic alcoholism, leads to diencephalic damage, hippocampal dysfunction, and spatial learning and memory deficits. We show a decrease in the magnitude of long-term potentiation (LTP) and paired-pulse facilitation (PPF) at CA3–CA1 synapses, independent of sex, following diencephalic damage induced by TD in rats. Thus, despite a lack of extensive hippocampal cell loss, diencephalic brain damage down-regulates plastic processes within the hippocampus, likely contributing to impaired hippocampal-dependent behaviors. However, both measures of hippocampal plasticity (LTP, PPF) were restored with brain-derived neurotrophic factor (BDNF), revealing an avenue for neural and behavioral recovery following diencephalic damage.
Collapse
Affiliation(s)
- Lindsey C Vedder
- Department of Psychology, Behavioral Neuroscience Program, Binghamton University, State University of New York, Binghamton, New York 13902, USA
| | - Lisa M Savage
- Department of Psychology, Behavioral Neuroscience Program, Binghamton University, State University of New York, Binghamton, New York 13902, USA
| |
Collapse
|
29
|
Bal-Price A, Lein PJ, Keil KP, Sethi S, Shafer T, Barenys M, Fritsche E, Sachana M, Meek MEB. Developing and applying the adverse outcome pathway concept for understanding and predicting neurotoxicity. Neurotoxicology 2016; 59:240-255. [PMID: 27212452 DOI: 10.1016/j.neuro.2016.05.010] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 05/13/2016] [Accepted: 05/13/2016] [Indexed: 12/12/2022]
Abstract
The Adverse Outcome Pathway (AOP) concept has recently been proposed to support a paradigm shift in regulatory toxicology testing and risk assessment. This concept is similar to the Mode of Action (MOA), in that it describes a sequence of measurable key events triggered by a molecular initiating event in which a stressor interacts with a biological target. The resulting cascade of key events includes molecular, cellular, structural and functional changes in biological systems, resulting in a measurable adverse outcome. Thereby, an AOP ideally provides information relevant to chemical structure-activity relationships as a basis for predicting effects of structurally similar compounds. AOPs could potentially also form the basis for qualitative and quantitative predictive modeling of the human adverse outcome resulting from molecular initiating or other key events for which higher-throughput testing methods are available or can be developed. A variety of cellular and molecular processes are known to be critical for normal function of the central (CNS) and peripheral nervous systems (PNS). Because of the biological and functional complexity of the CNS and PNS, it has been challenging to establish causative links and quantitative relationships between key events that comprise the pathways leading from chemical exposure to an adverse outcome in the nervous system. Following introduction of the principles of MOA and AOPs, examples of potential or putative adverse outcome pathways specific for developmental or adult neurotoxicity are summarized and aspects of their assessment considered. Their possible application in developing mechanistically informed Integrated Approaches to Testing and Assessment (IATA) is also discussed.
Collapse
Affiliation(s)
- Anna Bal-Price
- European Commission Joint Research Centre, Institute for Health and Consumer Protection, Ispra, Italy.
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA
| | - Kimberly P Keil
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA
| | - Sunjay Sethi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA
| | - Timothy Shafer
- Integrated Systems Toxicology Division, Office of Research and Development, U.S. Environmental Protection Agency, RTP, USA
| | - Marta Barenys
- IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Ellen Fritsche
- IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Magdalini Sachana
- European Commission Joint Research Centre, Institute for Health and Consumer Protection, Ispra, Italy
| | - M E Bette Meek
- McLaughlin Centre for Risk Science, University of Ottawa, Ottawa, Canada
| |
Collapse
|
30
|
Scholz-Starke J, Cesca F. Stepping Out of the Shade: Control of Neuronal Activity by the Scaffold Protein Kidins220/ARMS. Front Cell Neurosci 2016; 10:68. [PMID: 27013979 PMCID: PMC4789535 DOI: 10.3389/fncel.2016.00068] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 03/01/2016] [Indexed: 12/31/2022] Open
Abstract
The correct functioning of the nervous system depends on the exquisitely fine control of neuronal excitability and synaptic plasticity, which relies on an intricate network of protein-protein interactions and signaling that shapes neuronal homeostasis during development and in adulthood. In this complex scenario, Kinase D interacting substrate of 220 kDa/ankyrin repeat-rich membrane spanning (Kidins220/ARMS) acts as a multi-functional scaffold protein with preferential expression in the nervous system. Engaged in a plethora of interactions with membrane receptors, cytosolic signaling components and cytoskeletal proteins, Kidins220/ARMS is implicated in numerous cellular functions including neuronal survival, neurite outgrowth and maturation and neuronal activity, often in the context of neurotrophin (NT) signaling pathways. Recent studies have highlighted a number of cell- and context-specific roles for this protein in the control of synaptic transmission and neuronal excitability, which are at present far from being completely understood. In addition, some evidence has began to emerge, linking alterations of Kidins220 expression to the onset of various neurodegenerative diseases and neuropsychiatric disorders. In this review, we present a concise summary of our fragmentary knowledge of Kidins220/ARMS biological functions, focusing on the mechanism(s) by which it controls various aspects of neuronal activity. We have tried, where possible, to discuss the available evidence in the wider context of NT-mediated regulation, and to outline emerging roles of Kidins220/ARMS in human pathologies.
Collapse
Affiliation(s)
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia Genova, Italy
| |
Collapse
|
31
|
Eckenstaler R, Lessmann V, Brigadski T. CAPS1 effects on intragranular pH and regulation of BDNF release from secretory granules in hippocampal neurons. J Cell Sci 2016; 129:1378-90. [PMID: 26869227 DOI: 10.1242/jcs.178251] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 02/08/2016] [Indexed: 01/19/2023] Open
Abstract
The secretory protein brain-derived neurotrophic factor (BDNF) is assumed to be a key factor for the induction of synaptic plasticity processes in neurons. However, the molecular mechanisms for activity-dependent release of the protein largely remain elusive. Here, we demonstrate the relevance of the priming factor CAPS1 (also known as CADPS) for the maturation and exocytosis of BDNF-containing secretory granules, as well as for neurotransmitter release from synaptic vesicles. Using live-cell imaging and RNA silencing methods, we show that CAPS1 has a previously unrecognized function in regulating the intragranular pH of BDNF-containing secretory granules. Furthermore, our results demonstrate that acute single-cell knockdown of CAPS1 with unaltered expression in neighboring neurons leads to a strong reduction in the number of fusion-competent secretory granules and to a significant decrease of released BDNF following exocytosis in dendrites of CAPS1-deficient neurons. In addition, our results show a reduction in synaptic vesicle turnover after CAPS1 knockdown without affecting the density of active boutons in hippocampal neurons. Thus, our results reveal new functions of endogenous CAPS1 in the BDNF secretory granule life cycle, thereby representing a new mechanism of neuronal plasticity.
Collapse
Affiliation(s)
- Robert Eckenstaler
- Institute of Physiology, Medical Faculty, Otto-von-Guericke-University, Magdeburg 39120, Germany
| | - Volkmar Lessmann
- Institute of Physiology, Medical Faculty, Otto-von-Guericke-University, Magdeburg 39120, Germany Center of Behavioral Brain Sciences (CBBS), Magdeburg 39120, Germany
| | - Tanja Brigadski
- Institute of Physiology, Medical Faculty, Otto-von-Guericke-University, Magdeburg 39120, Germany Center of Behavioral Brain Sciences (CBBS), Magdeburg 39120, Germany
| |
Collapse
|
32
|
Amyloid-Beta Induced Changes in Vesicular Transport of BDNF in Hippocampal Neurons. Neural Plast 2016; 2016:4145708. [PMID: 26881108 PMCID: PMC4736975 DOI: 10.1155/2016/4145708] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/26/2015] [Accepted: 11/29/2015] [Indexed: 12/15/2022] Open
Abstract
The neurotrophin brain derived neurotrophic factor (BDNF) is an important growth factor in the CNS. Deficits in transport of this secretory protein could underlie neurodegenerative diseases. Investigation of disease-related changes in BDNF transport might provide insights into the cellular mechanism underlying, for example, Alzheimer's disease (AD). To analyze the role of BDNF transport in AD, live cell imaging of fluorescently labeled BDNF was performed in hippocampal neurons of different AD model systems. BDNF and APP colocalized with low incidence in vesicular structures. Anterograde as well as retrograde transport of BDNF vesicles was reduced and these effects were mediated by factors released from hippocampal neurons into the extracellular medium. Transport of BDNF was altered at a very early time point after onset of human APP expression or after acute amyloid-beta(1-42) treatment, while the activity-dependent release of BDNF remained unaffected. Taken together, extracellular cleavage products of APP induced rapid changes in anterograde and retrograde transport of BDNF-containing vesicles while release of BDNF was unaffected by transgenic expression of mutated APP. These early transport deficits might lead to permanently impaired brain functions in the adult brain.
Collapse
|
33
|
Edelmann E, Cepeda-Prado E, Franck M, Lichtenecker P, Brigadski T, Leßmann V. Theta Burst Firing Recruits BDNF Release and Signaling in Postsynaptic CA1 Neurons in Spike-Timing-Dependent LTP. Neuron 2015; 86:1041-1054. [DOI: 10.1016/j.neuron.2015.04.007] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Revised: 01/25/2015] [Accepted: 03/31/2015] [Indexed: 01/28/2023]
|
34
|
Kolarow R, Kuhlmann CRW, Munsch T, Zehendner C, Brigadski T, Luhmann HJ, Lessmann V. BDNF-induced nitric oxide signals in cultured rat hippocampal neurons: time course, mechanism of generation, and effect on neurotrophin secretion. Front Cell Neurosci 2014; 8:323. [PMID: 25426021 PMCID: PMC4224130 DOI: 10.3389/fncel.2014.00323] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 09/26/2014] [Indexed: 11/13/2022] Open
Abstract
BDNF and nitric oxide signaling both contribute to plasticity at glutamatergic synapses. However, the role of combined signaling of both pathways at the same synapse is largely unknown. Using NO imaging with diaminofluoresceine in cultured hippocampal neurons we analyzed the time course of neurotrophin-induced NO signals. Application of exogenous BDNF, NT-4, and NT-3 (but not NGF) induced NO signals in the soma and in proximal dendrites of hippocampal neurons that were sensitive to NO synthase activity, TrkB signaling, and intracellular calcium elevation. The effect of NO signaling on neurotrophin secretion was analyzed in BDNF-GFP, and NT-3-GFP transfected hippocampal neurons. Exogenous application of the NO donor sodium-nitroprusside markedly inhibited neurotrophin secretion. However, endogenously generated NO in response to depolarization and neurotrophin stimulation, both did not result in a negative feedback on neurotrophin secretion. These results suggest that a negative feedback of NO signaling on synaptic secretion of neurotrophins operates only at high intracellular levels of nitric oxide that are under physiological conditions not reached by depolarization or BDNF signaling.
Collapse
Affiliation(s)
- Richard Kolarow
- Medical Faculty, Institute of Physiology, Otto-von-Guericke-University Magdeburg, Germany ; University Medical Center, Institute of Physiology, Johannes Gutenberg-University Mainz Mainz, Germany
| | - Christoph R W Kuhlmann
- University Medical Center, Institute of Physiology, Johannes Gutenberg-University Mainz Mainz, Germany
| | - Thomas Munsch
- Medical Faculty, Institute of Physiology, Otto-von-Guericke-University Magdeburg, Germany
| | - Christoph Zehendner
- University Medical Center, Institute of Physiology, Johannes Gutenberg-University Mainz Mainz, Germany
| | - Tanja Brigadski
- Medical Faculty, Institute of Physiology, Otto-von-Guericke-University Magdeburg, Germany ; University Medical Center, Institute of Physiology, Johannes Gutenberg-University Mainz Mainz, Germany
| | - Heiko J Luhmann
- University Medical Center, Institute of Physiology, Johannes Gutenberg-University Mainz Mainz, Germany
| | - Volkmar Lessmann
- Medical Faculty, Institute of Physiology, Otto-von-Guericke-University Magdeburg, Germany ; University Medical Center, Institute of Physiology, Johannes Gutenberg-University Mainz Mainz, Germany
| |
Collapse
|
35
|
Martin JL, Finsterwald C. Cooperation between BDNF and glutamate in the regulation of synaptic transmission and neuronal development. Commun Integr Biol 2014. [DOI: 10.4161/cib.13761] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
36
|
Mendoza E, Miranda-Barrientos J, Vázquez-Roque R, Morales-Herrera E, Ruelas A, De la Rosa G, Flores G, Hernández-Echeagaray E. In vivo mitochondrial inhibition alters corticostriatal synaptic function and the modulatory effects of neurotrophins. Neuroscience 2014; 280:156-70. [DOI: 10.1016/j.neuroscience.2014.09.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 09/09/2014] [Indexed: 01/09/2023]
|
37
|
Montalbano A, Baj G, Papadia D, Tongiorgi E, Sciancalepore M. Blockade of BDNF signaling turns chemically-induced long-term potentiation into long-term depression. Hippocampus 2013; 23:879-89. [PMID: 23674394 DOI: 10.1002/hipo.22144] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2013] [Indexed: 01/28/2023]
Abstract
Long-term potentiation (LTP) is accompanied by increased spine density and dimensions triggered by signaling cascades involving activation of the neurotrophin brain-derived neurotrophic factor (BDNF) and cytoskeleton remodeling. Chemically-induced long-term potentiation (c-LTP) is a widely used cellular model of plasticity, whose effects on spines have been poorly investigated. We induced c-LTP by bath-application of the N-methyl-d-aspartate receptor (NMDAR) coagonist glycine or by the K(+) channel blocker tetraethylammonium (TEA) chloride in cultured hippocampal neurons and compared the changes in dendritic spines induced by the two models of c-LTP and determined if they depend on BDNF/TrkB signaling. We found that both TEA and glycine induced a significant increase in stubby spine density in primary and secondary apical dendrites, whereas a specific increase in mushroom spine density was observed upon TEA application only in primary dendrites. Both TEA and glycine increased BDNF levels and the blockade of tropomyosin-receptor-kinase receptors (TrkRs) by the nonselective tyrosine kinase inhibitor K-252a or the selective allosteric TrkB receptor (TrkBR) inhibitor ANA-12, abolished the c-LTP-induced increase in spine density. Surprisingly, a blockade of TrkBRs did not change basal spontaneous glutamatergic transmission but completely changed the synaptic plasticity induced by c-LTP, provoking a shift from a long-term increase to a long-term depression (LTD) in miniature excitatory postsynaptic current (mEPSC) frequency. In conclusion, these results suggest that BDNF/TrkB signaling is necessary for c-LTP-induced plasticity in hippocampal neurons and its blockade leads to a switch of c-LTP into chemical-LTD (c-LTD).
Collapse
Affiliation(s)
- A Montalbano
- Department of Life Sciences and B.R.A.I.N., Centre for Neuroscience, University of Trieste, Trieste, Italy
| | | | | | | | | |
Collapse
|
38
|
Leal G, Comprido D, Duarte CB. BDNF-induced local protein synthesis and synaptic plasticity. Neuropharmacology 2013; 76 Pt C:639-56. [PMID: 23602987 DOI: 10.1016/j.neuropharm.2013.04.005] [Citation(s) in RCA: 484] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Revised: 03/25/2013] [Accepted: 04/03/2013] [Indexed: 12/16/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is an important regulator of synaptic transmission and long-term potentiation (LTP) in the hippocampus and in other brain regions, playing a role in the formation of certain forms of memory. The effects of BDNF in LTP are mediated by TrkB (tropomyosin-related kinase B) receptors, which are known to be coupled to the activation of the Ras/ERK, phosphatidylinositol 3-kinase/Akt and phospholipase C-γ (PLC-γ) pathways. The role of BDNF in LTP is best studied in the hippocampus, where the neurotrophin acts at pre- and post-synaptic levels. Recent studies have shown that BDNF regulates the transport of mRNAs along dendrites and their translation at the synapse, by modulating the initiation and elongation phases of protein synthesis, and by acting on specific miRNAs. Furthermore, the effect of BDNF on transcription regulation may further contribute to long-term changes in the synaptic proteome. In this review we discuss the recent progress in understanding the mechanisms contributing to the short- and long-term regulation of the synaptic proteome by BDNF, and the role in synaptic plasticity, which is likely to influence learning and memory formation. This article is part of the Special Issue entitled 'BDNF Regulation of Synaptic Structure, Function, and Plasticity'.
Collapse
Affiliation(s)
- Graciano Leal
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Department of Life Sciences, University of Coimbra, 3004-517 Coimbra, Portugal
| | | | | |
Collapse
|
39
|
Brain-derived neurotrophic factor activation of CaM-kinase kinase via transient receptor potential canonical channels induces the translation and synaptic incorporation of GluA1-containing calcium-permeable AMPA receptors. J Neurosci 2012; 32:8127-37. [PMID: 22699894 DOI: 10.1523/jneurosci.6034-11.2012] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Glutamatergic synapses in early postnatal development transiently express calcium-permeable AMPA receptors (CP-AMPARs). Although these GluA2-lacking receptors are essential and are elevated in response to brain-derived neurotrophic factor (BDNF), little is known regarding molecular mechanisms that govern their expression and synaptic insertion. Here we show that BDNF-induced GluA1 translation in rat primary hippocampal neurons requires the activation of mammalian target of rapamycin (mTOR) via calcium calmodulin-dependent protein kinase kinase (CaMKK). Specifically, BDNF-mediated phosphorylation of threonine 308 (T308) in AKT, a known substrate of CaMKK and an upstream activator of mTOR-dependent translation, was prevented by (1) pharmacological inhibition of CaMKK with STO-609, (2) overexpression of a dominant-negative CaMKK, or (3) short hairpin-mediated knockdown of CaMKK. GluA1 surface expression induced by BDNF, as assessed by immunocytochemistry using an extracellular N-terminal GluA1 antibody or by surface biotinylation, was impaired following knockdown of CaMKK or treatment with STO-609. Activation of CaMKK by BDNF requires transient receptor potential canonical (TRPC) channels as SKF-96365, but not the NMDA receptor antagonist d-APV, prevented BDNF-induced GluA1 surface expression as well as phosphorylation of CaMKI, AKT(T308), and mTOR. Using siRNA we confirmed the involvement of TRPC5 and TRPC6 subunits in BDNF-induced AKT(T308) phosphorylation. The BDNF-induced increase in mEPSC was blocked by IEM-1460, a selected antagonist of CP-AMPARs, as well as by the specific repression of acute GluA1 translation via siRNA to GluA1 but not GluA2. Together these data support the conclusion that newly synthesized GluA1 subunits, induced by BDNF, are readily incorporated into synapses where they enhance the expression of CP-AMPARs and synaptic strength.
Collapse
|
40
|
Site-specific synapsin I phosphorylation participates in the expression of post-tetanic potentiation and its enhancement by BDNF. J Neurosci 2012; 32:5868-79. [PMID: 22539848 DOI: 10.1523/jneurosci.5275-11.2012] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
A large amount of experimental evidence has highlighted the rapid changes in synaptic efficacy induced by high-frequency stimulation and BDNF at central excitatory synapses. We clarified the quantal mechanisms and the involvement of Synapsin I (SynI) phosphorylation in the expression of post-tetanic potentiation (PTP) and in its modulation by BDNF in mouse glutamatergic autapses. We found that PTP is associated with an elevation in the probability of release and a concomitant increase in the size of the readily releasable pool (RRP). The latter component was virtually absent in SynI knock-out (KO) neurons, which indeed displayed impaired PTP. PTP was fully rescued by the expression of wild-type SynI, but not of its dephosphomimetic mutants in the phosphorylation sites for cAMP-dependent protein kinase and Ca²⁺/calmodulin-dependent protein kinases I/II. BDNF potently enhanced PTP through a further increase in the RRP size, which was missing in SynI KO neurons. In these neurons, the BDNF-induced PTP enhancement was rescued by the expression of wild-type SynI, but not of its dephosphomimetic mutant at the mitogen-dependent protein kinase sites. The results indicate that the increase in RRP size necessary for the full expression of PTP, and its sensitivity to BDNF, involve phosphorylation of SynI at distinct sites, thus implicating SynI as an essential downstream effector for the expression of PTP and for its enhancement by BDNF.
Collapse
|
41
|
Long-term behavioral and NMDA receptor effects of young-adult corticosterone treatment in BDNF heterozygous mice. Neurobiol Dis 2012; 46:722-31. [PMID: 22426399 DOI: 10.1016/j.nbd.2012.03.015] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 02/07/2012] [Accepted: 03/01/2012] [Indexed: 12/27/2022] Open
Abstract
Psychiatric illnesses, such as schizophrenia, are most likely caused by an interaction between genetic predisposition and environmental factors, including stress during development. The neurotrophin, brain-derived neurotrophic factor (BDNF) has been implicated in this illness as BDNF levels are decreased in the brain of patients with schizophrenia. The aim of the present study was to assess the combined effect of reduced BDNF levels and postnatal stress, simulated by chronic young-adult treatment with the stress hormone, corticosterone. From 6 weeks of age, female and male BDNF heterozygous mice and their wild-type controls were chronically treated with corticosterone in their drinking water for 3 weeks. At 11 weeks of age, male, but not female BDNF heterozygous mice treated with corticosterone exhibited a profound memory deficit in the Y-maze. There were no differences between the groups in baseline prepulse inhibition (PPI), a measure of sensorimotor gating, or its disruption by treatment with MK-801. However, an increase in startle caused by MK-801 treatment was absent in male, but not female BDNF heterozygous mice, irrespective of corticosterone treatment. Analysis of protein levels of the NMDA receptor subunits NR1, NR2A, NR2B and NR2C, showed a marked increase of NR2B levels in the dorsal hippocampus of male BDNF heterozygous mice treated with corticosterone. In the ventral hippocampus, significantly reduced levels of NR2A, NR2B and NR2C were observed in male BDNF heterozygous mice. The NMDA receptor effects in hippocampal sub-regions could be related to the spatial memory deficits and the loss of the effect of MK-801 on startle in these mice, respectively. No significant changes in NMDA receptor subunit levels were observed in any of the female groups. Similarly, no significant changes in levels of BDNF or its receptor, TrkB, were found other than the expected reduced levels of BDNF in heterozygous mice. In conclusion, the data show differential interactive effects of reduced levels of BDNF expression and corticosterone treatment on spatial memory and startle in male and female mice, accompanied by significant, but region-specific changes in NMDA receptor subunit levels in the dorsal and ventral hippocampus. These results could be important for our understanding of the interaction of neurodevelopmental stress and BDNF deficiency in cognitive and anxiety-related symptoms of psychiatric illnesses, such as schizophrenia.
Collapse
|
42
|
Favalli G, Li J, Belmonte-de-Abreu P, Wong AHC, Daskalakis ZJ. The role of BDNF in the pathophysiology and treatment of schizophrenia. J Psychiatr Res 2012; 46:1-11. [PMID: 22030467 DOI: 10.1016/j.jpsychires.2011.09.022] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2011] [Revised: 09/12/2011] [Accepted: 09/29/2011] [Indexed: 12/20/2022]
Abstract
Brain derived neurotrophic factor (BDNF) has been associated with the pathophysiology of schizophrenia (SCZ). However, it remains unclear whether alterations in BDNF observed in patients with SCZ are a core part of disease neurobiology or a consequence of treatment. In this manuscript we review existing knowledge relating the function of BDNF to synaptic transmission and neural plasticity and the relationship between BDNF and both pharmacological and non-pharmacological treatments for SCZ. With regards to synaptic transmission, exposure to BDNF or lack of this neurotrophin results in alteration to both excitatory and inhibitory synapses. Many authors have also evaluated the effects of both pharmacological and non-pharmacological treatments for SCZ in BDNF and despite some controversial results, it seems that medicated and non-medicated patients present with lower levels of BDNF when compared to controls. Further data suggests that typical antipsychotics may decrease BDNF expression whereas mixed results have been obtained with atypical antipsychotics. The authors found few studies reporting changes in BDNF after non-pharmacological treatments for SCZ, so the existing evidence in this area is limited. Although the study of BDNF provides some new insights into understanding of the pathophysiology and treatment of SCZ, additional work in this area is needed.
Collapse
|
43
|
Lauri S, Taira T. Kainate receptors in developing presynaptic terminals. ACTA ACUST UNITED AC 2011. [DOI: 10.1002/wmts.3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
44
|
Nathan PJ, Cobb SR, Lu B, Bullmore ET, Davies CH. Studying synaptic plasticity in the human brain and opportunities for drug discovery. Curr Opin Pharmacol 2011; 11:540-8. [DOI: 10.1016/j.coph.2011.06.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 06/15/2011] [Accepted: 06/15/2011] [Indexed: 12/15/2022]
|
45
|
Martin JL, Finsterwald C. Cooperation between BDNF and glutamate in the regulation of synaptic transmission and neuronal development. Commun Integr Biol 2011; 4:14-6. [PMID: 21509169 DOI: 10.4161/cib.4.1.13761] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Accepted: 09/27/2010] [Indexed: 01/06/2023] Open
Abstract
Ample evidence supports a role of brain-derived neurotrophic factor (BDNF) in the survival and differentiation of selective populations of neurons in the peripheral and central nervous systems. In addition to its trophic actions, BDNF exerts acute effects on synaptic transmission and plasticity. In particular, BDNF enhances excitatory synaptic transmission through pre- and postsynaptic mechanisms. In this regard, BDNF enhances glutamate release, the frequency of miniature excitatory postsynaptic currents (mEPSCs), NMDA receptor activity and the phosphorylation of NMDA receptor subunits. Our recent studies revealed a novel cooperative interaction between BDNF and glutamate in the regulation of dendritic development. Indeed, we found that the effects of BDNF on dendritic growth of cortical neurons require both the stimulation of cAMP response element-binding protein (CREB) phosphorylation by BDNF and the activation of the CREB-regulated transcription coactivator 1 (CRTC1) by glutamate. Together, these studies highlight the importance of the cooperation between BDNF and glutamate in the regulation of synaptic transmission and neuronal development.
Collapse
Affiliation(s)
- Jean-Luc Martin
- Department of Physiology; University of Lausanne; Lausanne, Switzerland
| | | |
Collapse
|
46
|
Lessmann V, Stroh-Kaffei S, Steinbrecher V, Edelmann E, Brigadski T, Kilb W, Luhmann HJ. The expression mechanism of the residual LTP in the CA1 region of BDNF k.o. mice is insensitive to NO synthase inhibition. Brain Res 2011; 1391:14-23. [PMID: 21458431 DOI: 10.1016/j.brainres.2011.03.061] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Revised: 03/23/2011] [Accepted: 03/24/2011] [Indexed: 01/22/2023]
Abstract
BDNF and nitric oxide signaling both contribute to long-term potentiation (LTP) at glutamatergic synapses, but to date, few studies analyzed the interaction of both signaling cascades in the same synaptic pathway. Here we addressed the question whether the residual LTP in the CA1 region of hippocampal slices from heterozygous BDNF knockout mice (BDNF⁺/⁻) is dependent on nitric oxide (NO) signaling. Extracellular recording of synaptic field potentials elicited by presynaptic Schaffer collateral stimulation was performed in the CA1 region of hippocampal slices of 4- to 6-week-old mice, and LTP was induced by a theta burst stimulation protocol. Application of the nitric oxide inhibitor L-NAME (200 μM) strongly inhibited LTP by 70% in wildtype animals. This inhibition of LTP was not a consequence of altered basal synaptic properties. In CA1 of BDNF⁺/⁻ mice, stimulated with the same theta burst protocol, LTP was reduced by 50% as compared to wildtype animals. This impairment in the expression of LTP in BDNF⁺/⁻ mice did not result from an increased synaptic fatigue. The residual LTP in BDNF⁺/⁻ was not further reduced by preincubation of slices with L-NAME. These results suggest that BDNF and NO share overlapping intracellular signaling cascades to mediate LTP in CA1, and part of their signaling cascades are most likely arranged consecutively in the signaling pathway mediating LTP.
Collapse
Affiliation(s)
- Volkmar Lessmann
- Institute of Physiology and Pathophysiology, University Medical Center Mainz, Johannes Gutenberg-University, Mainz, Germany.
| | | | | | | | | | | | | |
Collapse
|
47
|
Postsynaptic GluA1 enables acute retrograde enhancement of presynaptic function to coordinate adaptation to synaptic inactivity. Proc Natl Acad Sci U S A 2010; 107:21806-11. [PMID: 21098665 DOI: 10.1073/pnas.1016399107] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Prolonged blockade of AMPA-type glutamate receptors in hippocampal neuron cultures leads to homeostatic enhancements of pre- and postsynaptic function that appear correlated at individual synapses, suggesting some form of transsynaptic coordination. The respective modifications are important for overall synaptic strength but their interrelationship, dynamics, and molecular underpinnings are unclear. Here we demonstrate that adaptation begins postsynaptically but is ultimately communicated to presynaptic terminals and expressed as an accelerated turnover of synaptic vesicles. Critical postsynaptic modifications occur over hours, but enable retrograde communication within minutes once AMPA receptor (AMPAR) blockade is removed, causing elevation of both spontaneous and evoked vesicle fusion. The retrograde signaling does not require spiking activity and can be interrupted by NBQX, philanthotoxin, postsynaptic BAPTA, or external sequestration of BDNF, consistent with the acute release of retrograde messenger, triggered by postsynaptic Ca(2+) elevation via Ca(2+)-permeable AMPARs.
Collapse
|
48
|
Casalbore P, Barone I, Felsani A, D'Agnano I, Michetti F, Maira G, Cenciarelli C. Neural stem cells modified to express BDNF antagonize trimethyltin-induced neurotoxicity through PI3K/Akt and MAP kinase pathways. J Cell Physiol 2010; 224:710-721. [PMID: 20432466 DOI: 10.1002/jcp.22170] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In vitro expansion of neural stem cells (NSC) lentivirally transduced with human BDNF may serve as better cellular source for replacing degenerating neurons in disease, trauma and toxic insults. In this study, we evaluate the functional role of forced BDNF expression by means of NSC (M3GFP-BDNF) obtained from cerebral cortex of 1-day-old mice respect to NSC-control (M3GFP). We find that M3GFP-BDNF induced to differentiate significantly accumulate BDNF and undergone to high potassium-mediated depolarization, show rapid BDNF recycle and activation of Trk receptors signaling. Differentiated M3GFP-BDNF exhibit neurons and oligodendrocytes with extended processes although quantitative analyses of NSC-derived cell lineages show none statistical significance between both cell populations. Moreover, those cells show a significant induction of neuronal and oligodendroglial markers by RT-PCR and Western blot respect to M3GFP, such as betaIII-Tubulin, microtubule associated protein 2 (MAP2), neurofilaments heavy (NF-H), oligodendroglial myelin glycoprotein (OMG) and some molecules involved in glutamatergic synapse maturation, such as receptors tyrosine kinases (TRKs), post-synaptic density (PSD-95) and N-methyl-D-aspartate receptors 2 A/B (NMDA2A/B). After treatment with the neurotoxicant trimethyltin (TMT), differentiated M3GFP-BDNF exhibit an attenuation of cellular damage which correlates with a significant activation of MAPK and PI3K/Akt signaling and delayed activation of death signals, while on M3GFP, TMT induces a significant reduction of cell survival, neuronal differentiation and concomitant earlier activation of cleaved caspase-3. We demonstrate that overexpression of BDNF firmly regulate cell survival and differentiation of NSC and protects differentiated NSC against TMT-induced neurotoxicity through the PI3K/Akt and MAPK signaling pathways.
Collapse
Affiliation(s)
- Patrizia Casalbore
- Institute of Neurobiology and Molecular Medicine, National Research Council, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
49
|
|
50
|
Brain-derived neurotrophic factor controls activity-dependent maturation of CA1 synapses by downregulating tonic activation of presynaptic kainate receptors. J Neurosci 2009; 29:11294-303. [PMID: 19741136 DOI: 10.1523/jneurosci.0560-09.2009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Immature hippocampal synapses express presynaptic kainate receptors (KARs), which tonically inhibit glutamate release. Presynaptic maturation involves activity-dependent downregulation of the tonic KAR activity and consequent increase in release probability; however, the molecular mechanisms underlying this developmental process are unknown. Here, we have investigated whether brain derived neurotrophic factor (BDNF), a secreted protein implicated in developmental plasticity in several areas of the brain, controls presynaptic maturation by regulating KARs. Application of BDNF in neonate hippocampal slices resulted in increase in synaptic transmission that fully occluded the immature-type KAR activity in area CA1. Conversely, genetic ablation of BDNF was associated with delayed synaptic maturation and persistent presynaptic KAR activity, suggesting a role for endogenous BDNF in the developmental regulation of KAR function. In addition, our data suggests a critical role for BDNF TrkB signaling in fast activity-dependent regulation of KARs. Selective acute inhibition of TrkB receptors using a chemical-genetic approach prevented rapid change in synapse dynamics and loss of tonic KAR activity that is typically seen in response to induction of LTP at immature synapses. Together, these data show that BDNF-TrkB-dependent maturation of glutamatergic synapses is tightly associated with a loss of endogenous KAR activity. The coordinated action of these two receptor mechanisms has immediate physiological relevance in controlling presynaptic efficacy and transmission dynamics at CA3-CA1 synapses at a stage of development when functional contact already exists but transmission is weak.
Collapse
|