1
|
Mao LM, Thallapureddy K, Wang JQ. Effects of propofol on presynaptic synapsin phosphorylation in the mouse brain in vivo. Brain Res 2024; 1823:148671. [PMID: 37952872 PMCID: PMC10806815 DOI: 10.1016/j.brainres.2023.148671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/24/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023]
Abstract
The commonly used general anesthetic propofol can enhance the γ-aminobutyric acid-mediated inhibitory synaptic transmission and depress the glutamatergic excitatory synaptic transmission to achieve general anesthesia and other outcomes. In addition to the actions at postsynaptic sites, the modulation of presynaptic activity by propofol is thought to contribute to neurophysiological effects of the anesthetic, although potential targets of propofol within presynaptic nerve terminals are incompletely studied at present. In this study, we explored the possible linkage of propofol to synapsins, a family of neuron-specific phosphoproteins which are the most abundant proteins on presynaptic vesicles, in the adult mouse brain in vivo. We found that an intraperitoneal injection of propofol at a dose that caused loss of righting reflex increased basal levels of synapsin phosphorylation at the major representative phosphorylation sites (serine 9, serine 62/67, and serine 603) in the prefrontal cortex (PFC) of male and female mice. Propofol also elevated synapsin phosphorylation at these sites in the striatum and S9 and S62/67 phosphorylation in the hippocampus, while propofol had no effect on tyrosine hydroxylase phosphorylation in striatal nerve terminals. Total synapsin protein expression in the PFC, hippocampus, and striatum was not altered by propofol. These results reveal that synapsin could be a novel substrate of propofol in the presynaptic neurotransmitter release machinery. Propofol possesses the ability to upregulate synapsin phosphorylation in broad mouse brain regions.
Collapse
Affiliation(s)
- Li-Min Mao
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - Khyathi Thallapureddy
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - John Q Wang
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA; Department of Anesthesiology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA.
| |
Collapse
|
2
|
Serrano ME, Kim E, Petrinovic MM, Turkheimer F, Cash D. Imaging Synaptic Density: The Next Holy Grail of Neuroscience? Front Neurosci 2022; 16:796129. [PMID: 35401097 PMCID: PMC8990757 DOI: 10.3389/fnins.2022.796129] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/15/2022] [Indexed: 12/19/2022] Open
Abstract
The brain is the central and most complex organ in the nervous system, comprising billions of neurons that constantly communicate through trillions of connections called synapses. Despite being formed mainly during prenatal and early postnatal development, synapses are continually refined and eliminated throughout life via complicated and hitherto incompletely understood mechanisms. Failure to correctly regulate the numbers and distribution of synapses has been associated with many neurological and psychiatric disorders, including autism, epilepsy, Alzheimer’s disease, and schizophrenia. Therefore, measurements of brain synaptic density, as well as early detection of synaptic dysfunction, are essential for understanding normal and abnormal brain development. To date, multiple synaptic density markers have been proposed and investigated in experimental models of brain disorders. The majority of the gold standard methodologies (e.g., electron microscopy or immunohistochemistry) visualize synapses or measure changes in pre- and postsynaptic proteins ex vivo. However, the invasive nature of these classic methodologies precludes their use in living organisms. The recent development of positron emission tomography (PET) tracers [such as (18F)UCB-H or (11C)UCB-J] that bind to a putative synaptic density marker, the synaptic vesicle 2A (SV2A) protein, is heralding a likely paradigm shift in detecting synaptic alterations in patients. Despite their limited specificity, novel, non-invasive magnetic resonance (MR)-based methods also show promise in inferring synaptic information by linking to glutamate neurotransmission. Although promising, all these methods entail various advantages and limitations that must be addressed before becoming part of routine clinical practice. In this review, we summarize and discuss current ex vivo and in vivo methods of quantifying synaptic density, including an evaluation of their reliability and experimental utility. We conclude with a critical assessment of challenges that need to be overcome before successfully employing synaptic density biomarkers as diagnostic and/or prognostic tools in the study of neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Maria Elisa Serrano
- Department of Neuroimaging, The BRAIN Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| | - Eugene Kim
- Department of Neuroimaging, The BRAIN Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| | - Marija M Petrinovic
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Federico Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| | - Diana Cash
- Department of Neuroimaging, The BRAIN Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| |
Collapse
|
3
|
Yamaguchi H, Mano N. Analysis of membrane transport mechanisms of endogenous substrates using chromatographic techniques. Biomed Chromatogr 2019; 33:e4495. [PMID: 30661254 DOI: 10.1002/bmc.4495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 01/08/2019] [Accepted: 01/11/2019] [Indexed: 02/06/2023]
Abstract
Membrane transporters are expressed in various bodily tissues and play essential roles in the homeostasis of endogenous substances and the absortion, distribution and/or excretion of xenobiotics. For transporter assays, radioisotope-labeled compounds have been mainly used. However, commercially available radioisotope-labeled compounds are limited in number and relatively expensive. Chromatographic analyses such as high-performance liquid chromatography with ultraviolet absorptiometry and liquid chromatography with tandem mass spectrometry have also been applied for transport assays. To elucidate the transport properties of endogenous substrates, although there is no difficulty in performing assays using radioisotope-labeled probes, the endogenous background and the metabolism of the compound after its translocation across cell membranes must be considered when the intact compound is assayed. In this review, the current state of knowledge about the transport of endogenous substrates via membrane transporters as determined by chromatographic techniques is summarized. Chromatographic techniques have contributed to our understanding of the transport of endogenous substances including amino acids, catecholamines, bile acids, prostanoids and uremic toxins via membrane transporters.
Collapse
Affiliation(s)
- Hiroaki Yamaguchi
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan
| | - Nariyasu Mano
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan
| |
Collapse
|
4
|
Chen Z, Hu Q, Xie Q, Wu S, Pang Q, Liu M, Zhao Y, Tu F, Liu C, Chen X. Effects of Treadmill Exercise on Motor and Cognitive Function Recovery of MCAO Mice Through the Caveolin-1/VEGF Signaling Pathway in Ischemic Penumbra. Neurochem Res 2019; 44:930-946. [DOI: 10.1007/s11064-019-02728-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 01/09/2019] [Accepted: 01/10/2019] [Indexed: 11/29/2022]
|
5
|
Kim JH, Kim HJ, Yu DH, Kweon HS, Huh YH, Kim HR. Changes in numbers and size of synaptic vesicles of cortical neurons induced by exposure to 835 MHz radiofrequency-electromagnetic field. PLoS One 2017; 12:e0186416. [PMID: 29045446 PMCID: PMC5646811 DOI: 10.1371/journal.pone.0186416] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 09/29/2017] [Indexed: 12/17/2022] Open
Abstract
We studied the effects of radiofrequency electromagnetic fields (RF-EMFs) exposure on neuronal functions of mice. Particularly, we focused on RF-EMF effects on synaptic vesicles (SVs), which store neurotransmitters at axon terminals or synaptic boutons. C57 BL/6 mice were exposed to 835 MHz RF-EMF (4.0 W/kg SAR, for 5 h daily) and alterations in SVs at presynaptic terminals in the cerebral cortex were determined. Ultrastructure of randomly selected cortical neurons was observed using typical electron microscopy and bio-high voltage electron microscopy (Bio-HVEM) methods, which enable the estimation of the numbers and size of SVs. The density of the SVs (number /10 μm2 or 40 μm3) was significantly decreased in the presynaptic boutons of cortical neurons after RF-EMF exposure. Furthermore, qPCR and immunoblotting analyses revealed that the expression of synapsins I/II (Syns I/II) genes and proteins were significantly decreased in the cortical neurons of RF-EMF exposed mice. The present study suggested that alteration of SVs and Syn levels may result in alterations of neurotransmitters in the cerebral cortex following RF-EMF exposure.
Collapse
Affiliation(s)
- Ju Hwan Kim
- Department of Pharmacology, College of Medicine, Dankook University, Cheonan, Chungnam, South Korea
| | - Hyo-Jeong Kim
- Department of Pharmacology, College of Medicine, Dankook University, Cheonan, Chungnam, South Korea
- Center for Electron Microscopy Research, Korea Basic Science Institute, Ochang, Chungbuk, South Korea
| | - Da-Hyeon Yu
- Department of Pharmacology, College of Medicine, Dankook University, Cheonan, Chungnam, South Korea
| | - Hee-Seok Kweon
- Center for Electron Microscopy Research, Korea Basic Science Institute, Ochang, Chungbuk, South Korea
| | - Yang Hoon Huh
- Center for Electron Microscopy Research, Korea Basic Science Institute, Ochang, Chungbuk, South Korea
- * E-mail: (HRK); (YHH)
| | - Hak Rim Kim
- Department of Pharmacology, College of Medicine, Dankook University, Cheonan, Chungnam, South Korea
- * E-mail: (HRK); (YHH)
| |
Collapse
|
6
|
Validated methods for determination of neurotransmitters and metabolites in rodent brain tissue and extracellular fluid by reversed phase UHPLC–MS/MS. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1028:120-129. [DOI: 10.1016/j.jchromb.2016.06.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/03/2016] [Accepted: 06/06/2016] [Indexed: 11/19/2022]
|
7
|
Basu D, Tian Y, Hui P, Bhandari J, Johnson RL, Mishra RK. Change in expression of vesicular protein synapsin II by chronic treatment with D2 allosteric modulator PAOPA. Peptides 2015; 66:58-62. [PMID: 25703303 DOI: 10.1016/j.peptides.2015.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 01/20/2015] [Accepted: 01/20/2015] [Indexed: 12/21/2022]
Abstract
The hallmark symptoms of schizophrenia include profound disturbances in thought, perception, cognition etc., which negatively impacts an individual's quality of life. Current antipsychotic drugs are not effective in treating all symptoms of this disorder, and often cause severe movement and metabolic side effects. Consequently, there remains a strong impetus to develop safer and more efficacious therapeutics for patients, as well as elucidating the etiology of schizophrenia. Previous work in our lab has introduced a novel candidate for the treatment of this disease: the dopamine D2 receptor (D2R) allosteric modulator, 3(R)-[(2(S)-pyrrolidinylcarbonyl)amino]-2-oxo-1-pyrrolidineacetamide (PAOPA). We have previously shown that PAOPA, by selectively modulating D2R, can ameliorate schizophrenia-like symptoms in animal models, although the precise mechanism is presently not understood. Synapsin II is a presynaptic vesicular protein which has been strongly implicated in schizophrenia, as it is reduced in the prefrontal cortex of patients, and knockdown of this protein elicits schizophrenia-like phenotypes in animal models. Given the therapeutic effects of PAOPA and the role of synapsin II in schizophrenia, the objective of this study was to investigate the effect of chronic administration of PAOPA (45 days) on neuronal synapsin II protein expression in rodents. Immunoblot results revealed that the synapsin IIa, but not the IIb isoform, was increased in the dopaminergic regions of the striatum, nucleus accumbens, and medial prefrontal cortex. The results of this study implicate a role for modulation of synapsin II as a possible therapeutic mechanism of action for potential antipsychotic drug PAOPA.
Collapse
Affiliation(s)
- Dipannita Basu
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, 1200 Main St. W., Hamilton, ON, Canada L8N 3Z5
| | - Yuxin Tian
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, 1200 Main St. W., Hamilton, ON, Canada L8N 3Z5
| | - Patricia Hui
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, 1200 Main St. W., Hamilton, ON, Canada L8N 3Z5
| | - Jayant Bhandari
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, 1200 Main St. W., Hamilton, ON, Canada L8N 3Z5
| | - Rodney L Johnson
- Department of Medicinal Chemistry, University of Minnesota, 308 Harvard Street SE, Minneapolis, MN 55455, USA
| | - Ram K Mishra
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, 1200 Main St. W., Hamilton, ON, Canada L8N 3Z5.
| |
Collapse
|
8
|
Hong YP, Lee HC, Kim HT. Treadmill exercise after social isolation increases the levels of NGF, BDNF, and synapsin I to induce survival of neurons in the hippocampus, and improves depression-like behavior. J Exerc Nutrition Biochem 2015; 19:11-8. [PMID: 25960950 PMCID: PMC4424441 DOI: 10.5717/jenb.2015.19.1.11] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 02/01/2015] [Accepted: 02/05/2015] [Indexed: 11/16/2022] Open
Abstract
[Purpose] We investigated the effects of 8 weeks of treadmill exercise on nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and synapsin I protein expression and on the number of 5-bromo-2'-deoxyuridine-5'-mono-phosphate (BrdU)-positive cells in the dentate gyrus of the hippocampus in socially isolated rats. Additionally, we examined the effects of exercise on the number of serotonin (5-HT)- and tryptophan hydroxylase (TPH)-positive cells in the raphe nuclei and on depression behaviors induced by social isolation. [Methods] Forty male Sprague-Dawley rats were divided into four groups: (1) group housing and control group (GCG, n = 10); (2) group housing and exercise group (GEG, n = 10); (3) isolated housing and control group (ICG, n = 10); and (4) isolated housing and exercise group (IEG, n = 10). After 1 week of housing under the normal condition of 3 animals per cage, rats were socially isolated via transfer to individual cages for 8 weeks. Rats were then subjected to treadmill exercise for 5 days per week for 8 weeks during which time the speed of the treadmill was gradually increased. [Results] Compared to the GCG, levels of NGF, BDNF, and synapsin I were significantly decreased in the ICG and significantly increased in the IEG (p < 0.001 respectively). Significantly more BrdU-positive cells in the GEG were present as compared to the GCG and ICG, and more BrdU-positive cells were found in the IEG as compared to the ICG (p < 0.001). 5-HT-positive cells in the GEG were significantly increased compared to the GCG and ICG, and more of these cells were found in the IEG as compared to the ICG (p < 0.01). TPH-positive cells in the GEG were significantly increased compared to those in the GCG and ICG (p < 0.05). In the forced swim test, immobility time was significantly increased in the ICG and significantly decreased in the IEG as compared to the ICG (p < 0.01). [Conclusion] These results showed that regular treadmill exercise following social isolation not only increased the levels of NGF, BDNF, and synapsin I to induce survival of neurons in the hippocampus but also improved depression by increasing the number of serotonergic cells in the raphe nuclei.
Collapse
Affiliation(s)
- Young-Pyo Hong
- Department of Health and Sport Science, Korea National Sport University, Seoul, Korea
| | - Hyo-Chul Lee
- Department of Health and Sport Science, Korea National Sport University, Seoul, Korea
| | - Hyun-Tae Kim
- Department of Health and Sport Science, Korea National Sport University, Seoul, Korea
| |
Collapse
|
9
|
Sun WL, Eisenstein SA, Zelek-Molik A, McGinty JF. A single brain-derived neurotrophic factor infusion into the dorsomedial prefrontal cortex attenuates cocaine self-administration-induced phosphorylation of synapsin in the nucleus accumbens during early withdrawal. Int J Neuropsychopharmacol 2015; 18:pyu049. [PMID: 25522393 PMCID: PMC4368877 DOI: 10.1093/ijnp/pyu049] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Dysregulation in the prefrontal cortex-nucleus accumbens pathway has been implicated in cocaine addiction. We have previously demonstrated that one intra-dorsomedial prefrontal cortex brain-derived neurotrophic factor (BDNF) infusion immediately following the last cocaine self-administration session caused a long-lasting inhibition of cocaine-seeking and normalized the cocaine-induced disturbance of glutamate transmission in the nucleus accumbens after extinction and a cocaine prime. However, the molecular mechanism mediating the brain-derived neurotrophic factor effect on cocaine-induced alterations in extracellular glutamate levels is unknown. METHODS In the present study, we determined the effects of brain-derived neurotrophic factor on cocaine-induced changes in the phosphorylation of synapsin (p-synapsin), a family of presynaptic proteins that mediate synaptic vesicle mobilization, in the nucleus accumbens during early withdrawal. RESULTS Two hours after cocaine self-administration, p-synapsin Ser9 and p-synapsin Ser62/67, but not p-synapsin Ser603, were increased in the nucleus accumbens. At 22 hours, only p-synapsin Ser9 was still elevated. Elevations at both time points were attenuated by an intra-dorsomedial prefrontal cortex brain-derived neurotrophic factor infusion immediately after the end of cocaine self-administration. Brain-derived neurotrophic factor also reduced cocaine self-administration withdrawal-induced phosphorylation of the protein phosphatase 2A C-subunit, suggesting that brain-derived neurotrophic factor disinhibits protein phosphatase 2A C-subunit, consistent with p-synapsin Ser9 dephosphorylation. Further, co-immunoprecipitation demonstrated that protein phosphatase 2A C-subunit and synapsin are associated in a protein-protein complex that was reduced after 2 hours of withdrawal from cocaine self-administration and reversed by brain-derived neurotrophic factor. CONCLUSIONS Taken together, these findings demonstrate that brain-derived neurotrophic factor normalizes the cocaine self-administration-induced elevation of p-synapsin in nucleus accumbens that may underlie a disturbance in the probability of neurotransmitter release or represent a compensatory neuroadaptation in response to the hypofunction within the prefrontal cortex-nucleus accumbens pathway during cocaine withdrawal.
Collapse
Affiliation(s)
- Wei-Lun Sun
- Department of Neurosciences and Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina, (Drs Sun and McGinty); Department of Psychiatry, Washington University, St Louis, MO (Dr Eisenstein); Department of Brain Biochemistry, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland (Dr Zelek-Molik)
| | - Sarah A Eisenstein
- Department of Neurosciences and Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina, (Drs Sun and McGinty); Department of Psychiatry, Washington University, St Louis, MO (Dr Eisenstein); Department of Brain Biochemistry, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland (Dr Zelek-Molik)
| | - Agnieszka Zelek-Molik
- Department of Neurosciences and Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina, (Drs Sun and McGinty); Department of Psychiatry, Washington University, St Louis, MO (Dr Eisenstein); Department of Brain Biochemistry, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland (Dr Zelek-Molik)
| | - Jacqueline F McGinty
- Department of Neurosciences and Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina, (Drs Sun and McGinty); Department of Psychiatry, Washington University, St Louis, MO (Dr Eisenstein); Department of Brain Biochemistry, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland (Dr Zelek-Molik).
| |
Collapse
|
10
|
Yoon HH, Park JH, Kim YH, Min J, Hwang E, Lee CJ, Suh JKF, Hwang O, Jeon SR. Optogenetic inactivation of the subthalamic nucleus improves forelimb akinesia in a rat model of Parkinson disease. Neurosurgery 2014; 74:533-40; discussion 540-1. [PMID: 24463495 DOI: 10.1227/neu.0000000000000297] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The inhibition of neuronal activity by electrical deep brain stimulation is one of the mechanisms explaining the therapeutic effects in patients with Parkinson disease (PD) but cannot specifically activate or inactivate different types of neurons. Recently, a new technology based on optogenetics has been developed to modulate the activity of specific neurons. However, the therapeutic effects of optical inactivation in the subthalamic nucleus (STN) have not been fully investigated. OBJECTIVE To perform various behavioral tests to evaluate changes in motor functions in a PD rat model after optogene expression and, unlike previous studies, to assess the therapeutic effects of direct optogenetic inactivation in the STN. METHODS 6-Hydroxydopamine-induced hemiparkinsonian rats received injections of hSynapsin1-NpHR-YFP adeno-associated virus or an equivalent volume of phosphate-buffered saline. Three weeks after injection of adeno-associated virus or phosphate-buffered saline, the optic fiber was implanted into the ipsilateral STN. A stepping test, a cylinder test, and an apomorphine-induced rotation test were performed in 3 sequential steps: during light-off state, during light stimulation, and again during light-off state. RESULTS Stepping tests revealed that optical inhibition of the STN significantly improved 6-hydroxydopamine-induced forelimb akinesia. PD motor signs, as assessed by cylinder and apomorphine tests, were not affected by optical inhibition. Immunofluorescence revealed that halorhodopsin was highly expressed and colocalized with vesicular glutamate transporter 2 in the STN. CONCLUSION Optogenetic inhibition in the STN may be effective in improving contralateral forelimb akinesia but not in changing forelimb preference or reducing dopaminergic receptor supersensitivity. These findings are useful as a basis for future studies on optogenetics in PD.
Collapse
Affiliation(s)
- Hyung Ho Yoon
- ‡Department of Neurological Surgery and **Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, Republic of Korea; §Department of Neurological Surgery, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Republic of Korea; ¶Department of Computer Science and Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana; ‖Center for Neural Science and WCI Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul, Republic of Korea; #Center for Bionics of Korea Institute of Science and Technology, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Berglind F, Ledri M, Sørensen AT, Nikitidou L, Melis M, Bielefeld P, Kirik D, Deisseroth K, Andersson M, Kokaia M. Optogenetic inhibition of chemically induced hypersynchronized bursting in mice. Neurobiol Dis 2014; 65:133-41. [PMID: 24491965 DOI: 10.1016/j.nbd.2014.01.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 01/23/2014] [Indexed: 10/25/2022] Open
Abstract
Synchronized activity is common during various physiological operations but can culminate in seizures and consequently in epilepsy in pathological hyperexcitable conditions in the brain. Many types of seizures are not possible to control and impose significant disability for patients with epilepsy. Such intractable epilepsy cases are often associated with degeneration of inhibitory interneurons in the cortical areas resulting in impaired inhibitory drive onto the principal neurons. Recently emerging optogenetic technique has been proposed as an alternative approach to control such seizures but whether it may be effective in situations where inhibitory processes in the brain are compromised has not been addressed. Here we used pharmacological and optogenetic techniques to block inhibitory neurotransmission and induce epileptiform activity in vitro and in vivo. We demonstrate that NpHR-based optogenetic hyperpolarization and thereby inactivation of a principal neuronal population in the hippocampus is effectively attenuating seizure activity caused by disconnected network inhibition both in vitro and in vivo. Our data suggest that epileptiform activity in the hippocampus caused by impaired inhibition may be controlled by optogenetic silencing of principal neurons and potentially can be developed as an alternative treatment for epilepsy.
Collapse
Affiliation(s)
- Fredrik Berglind
- Experimental Epilepsy Group, Epilepsy Center, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden
| | - Marco Ledri
- Experimental Epilepsy Group, Epilepsy Center, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden
| | - Andreas Toft Sørensen
- Experimental Epilepsy Group, Epilepsy Center, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden
| | - Litsa Nikitidou
- Experimental Epilepsy Group, Epilepsy Center, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden
| | - Miriam Melis
- Experimental Epilepsy Group, Epilepsy Center, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden
| | - Pascal Bielefeld
- Experimental Epilepsy Group, Epilepsy Center, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden
| | - Deniz Kirik
- Brain Repair and Imaging in Neural Systems (BRAINS) Unit, Department of Experimental Medical Sciences, Lund University, SE-22184 Lund, Sweden
| | - Karl Deisseroth
- Dept. of Bioengineering, Stanford University, 94305 Stanford, CA, USA
| | - My Andersson
- Experimental Epilepsy Group, Epilepsy Center, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden
| | - Merab Kokaia
- Experimental Epilepsy Group, Epilepsy Center, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden.
| |
Collapse
|
12
|
Vasileva M, Renden R, Horstmann H, Gitler D, Kuner T. Overexpression of synapsin Ia in the rat calyx of Held accelerates short-term plasticity and decreases synaptic vesicle volume and active zone area. Front Cell Neurosci 2013; 7:270. [PMID: 24391547 PMCID: PMC3868894 DOI: 10.3389/fncel.2013.00270] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 12/04/2013] [Indexed: 01/10/2023] Open
Abstract
Synapsins are synaptic vesicle (SV) proteins organizing a component of the reserve pool of vesicles at most central nervous system synapses. Alternative splicing of the three mammalian genes results in multiple isoforms that may differentially contribute to the organization and maintenance of the SV pools. To address this, we first characterized the expression pattern of synapsin isoforms in the rat calyx of Held. At postnatal day 16, synapsins Ia, Ib, IIb and IIIa were present, while IIa-known to sustain repetitive transmission in glutamatergic terminals-was not detectable. To test if the synapsin I isoforms could mediate IIa-like effect, and if this depends on the presence of the E-domain, we overexpressed either synapsin Ia or synapsin Ib in the rat calyx of Held via recombinant adeno-associated virus-mediated gene transfer. Although the size and overall structure of the perturbed calyces remained unchanged, short-term depression and recovery from depression were accelerated upon overexpression of synapsin I isoforms. Using electron microscopic three-dimensional reconstructions we found a redistribution of SV clusters proximal to the active zones (AZ) alongside with a decrease of both AZ area and SV volume. The number of SVs at individual AZs was strongly reduced. Hence, our data indicate that the amount of synapsin Ia expressed in the calyx regulates the rate and extent of short-term synaptic plasticity by affecting vesicle recruitment to the AZ. Finally, our study reveals a novel contribution of synapsin Ia to define the surface area of AZs.
Collapse
Affiliation(s)
- Mariya Vasileva
- Institute of Anatomy and Cell Biology, Heidelberg University Heidelberg, Germany
| | - Robert Renden
- Institute of Anatomy and Cell Biology, Heidelberg University Heidelberg, Germany
| | - Heinz Horstmann
- Institute of Anatomy and Cell Biology, Heidelberg University Heidelberg, Germany
| | - Daniel Gitler
- Department of Physiology and Cell Biology, Faculty of Health Sciences and Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev Beer-Sheva, Israel
| | - Thomas Kuner
- Institute of Anatomy and Cell Biology, Heidelberg University Heidelberg, Germany
| |
Collapse
|
13
|
Wigestrand M, Stenberg M, Walaas S, Fonnum F, Andersson P. Non-dioxin-like PCBs inhibit [3H]WIN-35,428 binding to the dopamine transporter: A structure–activity relationship study. Neurotoxicology 2013; 39:18-24. [DOI: 10.1016/j.neuro.2013.07.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 07/24/2013] [Accepted: 07/24/2013] [Indexed: 11/29/2022]
|
14
|
Rodrigues HA, Fonseca MDC, Camargo WL, Lima PMA, Martinelli PM, Naves LA, Prado VF, Prado MAM, Guatimosim C. Reduced expression of the vesicular acetylcholine transporter and neurotransmitter content affects synaptic vesicle distribution and shape in mouse neuromuscular junction. PLoS One 2013; 8:e78342. [PMID: 24260111 PMCID: PMC3832638 DOI: 10.1371/journal.pone.0078342] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 09/18/2013] [Indexed: 12/03/2022] Open
Abstract
In vertebrates, nerve muscle communication is mediated by the release of the neurotransmitter acetylcholine packed inside synaptic vesicles by a specific vesicular acetylcholine transporter (VAChT). Here we used a mouse model (VAChT KDHOM) with 70% reduction in the expression of VAChT to investigate the morphological and functional consequences of a decreased acetylcholine uptake and release in neuromuscular synapses. Upon hypertonic stimulation, VAChT KDHOM mice presented a reduction in the amplitude and frequency of miniature endplate potentials, FM 1–43 staining intensity, total number of synaptic vesicles and altered distribution of vesicles within the synaptic terminal. In contrast, under electrical stimulation or no stimulation, VAChT KDHOM neuromuscular junctions did not differ from WT on total number of vesicles but showed altered distribution. Additionally, motor nerve terminals in VAChT KDHOM exhibited small and flattened synaptic vesicles similar to that observed in WT mice treated with vesamicol that blocks acetylcholine uptake. Based on these results, we propose that decreased VAChT levels affect synaptic vesicle biogenesis and distribution whereas a lower ACh content affects vesicles shape.
Collapse
Affiliation(s)
- Hermann A. Rodrigues
- Departamento de Morfologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brasil
| | - Matheus de C. Fonseca
- Departamento de Morfologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brasil
| | - Wallace L. Camargo
- Departamento de Fisiologia e Biofísica, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brasil
| | - Patrícia M. A. Lima
- Departamento de Engenharia de Biossistemas, Universidade Federal de São João Del Rei, São João Del Rei, Brasil
| | - Patrícia M. Martinelli
- Departamento de Morfologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brasil
| | - Lígia A. Naves
- Departamento de Fisiologia e Biofísica, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brasil
| | - Vânia F. Prado
- Robarts Research Institute and Department of Physiology and Pharmacology and Anatomy & Cell Biology, University of Western Ontario, London, ON, Canada
| | - Marco A. M. Prado
- Robarts Research Institute and Department of Physiology and Pharmacology and Anatomy & Cell Biology, University of Western Ontario, London, ON, Canada
| | - Cristina Guatimosim
- Departamento de Morfologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brasil
- * E-mail:
| |
Collapse
|
15
|
Lerner I, Bentin S, Shriki O. Excessive attractor instability accounts for semantic priming in schizophrenia. PLoS One 2012; 7:e40663. [PMID: 22844407 PMCID: PMC3402492 DOI: 10.1371/journal.pone.0040663] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 06/11/2012] [Indexed: 11/23/2022] Open
Abstract
One of the most pervasive findings in studies of schizophrenics with thought disorders is their peculiar pattern of semantic priming, which presumably reflects abnormal associative processes in the semantic system of these patients. Semantic priming is manifested by faster and more accurate recognition of a word-target when preceded by a semantically related prime, relative to an unrelated prime condition. Compared to control, semantic priming in schizophrenics is characterized by reduced priming effects at long prime-target Stimulus Onset Asynchrony (SOA) and, sometimes, augmented priming at short SOA. In addition, unlike controls, schizophrenics consistently show indirect (mediated) priming (such as from the prime ‘wedding’ to the target ‘finger’, mediated by ‘ring’). In a previous study, we developed a novel attractor neural network model with synaptic adaptation mechanisms that could account for semantic priming patterns in healthy individuals. Here, we examine the consequences of introducing attractor instability to this network, which is hypothesized to arise from dysfunctional synaptic transmission known to occur in schizophrenia. In two simulated experiments, we demonstrate how such instability speeds up the network’s dynamics and, consequently, produces the full spectrum of priming effects previously reported in patients. The model also explains the inconsistency of augmented priming results at short SOAs using directly related pairs relative to the consistency of indirect priming. Further, we discuss how the same mechanism could account for other symptoms of the disease, such as derailment (‘loose associations’) or the commonly seen difficulty of patients in utilizing context. Finally, we show how the model can statistically implement the overly-broad wave of spreading activation previously presumed to characterize thought-disorders in schizophrenia.
Collapse
Affiliation(s)
- Itamar Lerner
- Interdisciplinary Center for Neural Computation, The Hebrew University of Jerusalem, Jerusalem, Israel
- * E-mail: (OS); (IL)
| | - Shlomo Bentin
- Department of Psychology and Interdisciplinary Center for Neural Computation, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Oren Shriki
- Section on Critical Brain Dynamics, National Institute of Mental Health, Bethesda, Maryland, United States of America
- * E-mail: (OS); (IL)
| |
Collapse
|
16
|
Vasileva M, Horstmann H, Geumann C, Gitler D, Kuner T. Synapsin-dependent reserveo pool of synaptic vesicles supports replenishment of the readily releasable pool under intense synaptic transmission. Eur J Neurosci 2012; 36:3005-20. [DOI: 10.1111/j.1460-9568.2012.08225.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
17
|
Synapsin selectively controls the mobility of resting pool vesicles at hippocampal terminals. J Neurosci 2012; 32:3969-80. [PMID: 22442064 DOI: 10.1523/jneurosci.5058-11.2012] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Presynaptic terminals are specialized sites for information transmission where vesicles fuse with the plasma membrane and are locally recycled. Recent work has extended this classical view, with the observation that a subset of functional vesicles is dynamically shared between adjacent terminals by lateral axonal transport. Conceptually, such transport would be expected to disrupt vesicle retention around the active zone, yet terminals are characterized by a high-density vesicle cluster, suggesting that counteracting stabilizing mechanisms must operate against this tendency. The synapsins are a family of proteins that associate with synaptic vesicles and determine vesicle numbers at the terminal, but their specific function remains controversial. Here, using multiple quantitative fluorescence-based approaches and electron microscopy, we show that synapsin is instrumental for resisting vesicle dispersion and serves as a regulatory element for controlling lateral vesicle sharing between synapses. Deleting synapsin disrupts the organization of presynaptic vesicle clusters, making their boundaries hard to define. Concurrently, the fraction of vesicles amenable to transport is increased, and more vesicles are translocated to the axon. Importantly, in neurons from synapsin knock-out mice the resting and recycling pools are equally mobile. Synapsin, when present, specifically restricts the mobility of resting pool vesicles without affecting the division of vesicles between these pools. Specific expression of synapsin IIa, the sole isoform affecting synaptic depression, rescues the knock-out phenotype. Together, our results show that synapsin is pivotal for maintaining synaptic vesicle cluster integrity and that it contributes to the regulated sharing of vesicles between terminals.
Collapse
|
18
|
Wendt A, Speidel D, Danielsson A, Esguerra JLS, Bogen IL, Walaas SI, Salehi A, Eliasson L. Synapsins I and II are not required for insulin secretion from mouse pancreatic β-cells. Endocrinology 2012; 153:2112-9. [PMID: 22334712 DOI: 10.1210/en.2011-1702] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Synapsins are a family of phosphoproteins that modulate the release of neurotransmitters from synaptic vesicles. The release of insulin from pancreatic β-cells has also been suggested to be regulated by synapsins. In this study, we have utilized a knock out mouse model with general disruptions of the synapsin I and II genes [synapsin double knockout (DKO)]. Stimulation with 20 mm glucose increased insulin secretion 9-fold in both wild-type (WT) and synapsin DKO islets, whereas secretion in the presence of 70 mm K(+) and 1 mm glucose was significantly enhanced in the synapsin DKO mice compared to WT. Exocytosis in single β-cells was investigated using patch clamp. The exocytotic response, measured by capacitance measurements and elicited by a depolarization protocol designed to visualize exocytosis of vesicles from the readily releasable pool and from the reserve pool, was of the same size in synapsin DKO and WT β-cells. The increase in membrane capacitance corresponding to readily releasable pool was approximately 50fF in both genotypes. We next investigated the voltage-dependent Ca(2+) influx. In both WT and synapsin DKO β-cells the Ca(2+) current peaked at 0 mV and measured peak current (I(p)) and net charge (Q) were of similar magnitude. Finally, ultrastructural data showed no variation in total number of granules (N(v)) or number of docked granules (N(s)) between the β-cells from synapsin DKO mice and WT control. We conclude that neither synapsin I nor synapsin II are directly involved in the regulation of glucose-stimulated insulin secretion and Ca(2)-dependent exocytosis in mouse pancreatic β-cells.
Collapse
Affiliation(s)
- Anna Wendt
- Lund University Diabetes Centre, Department of Clinical Sciences in Malmö, Islet Cell Exocytosis, Lund University, 20502 Malmö, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Dyck BA, Beyaert MGR, Ferro MA, Mishra RK. Medial prefrontal cortical synapsin II knock-down induces behavioral abnormalities in the rat: examining synapsin II in the pathophysiology of schizophrenia. Schizophr Res 2011; 130:250-9. [PMID: 21689907 DOI: 10.1016/j.schres.2011.05.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Revised: 05/17/2011] [Accepted: 05/19/2011] [Indexed: 01/11/2023]
Abstract
Synapsin II is a synaptic vesicle-associated phosphoprotein that has been implicated in the pathophysiology of schizophrenia. Studies have demonstrated reductions in synapsin II mRNA and protein in medial prefrontal cortical post-mortem samples from patients with schizophrenia, genetic associations between synapsin II and schizophrenia, and synapsin II protein regulation by dopamine receptor activation. Collectively, this research indicates a relationship between synapsin II dysregulation and schizophrenia; however, it remains unknown whether perturbations in synapsin II play a role in the pathophysiology of this disease. The aim of this project was to evaluate animals with selective knock-down of synapsin II in the medial prefrontal cortex. After continuous infusion of synapsin II antisense sequences, animals were examined for the presence of schizophrenic-like behavioral phenotypes and assessed on the response to clinically relevant antipsychotic drugs. Our results indicate that rats with selective reductions in medial prefrontal cortical synapsin II demonstrate deficits in sensorimotor gating (prepulse inhibition), reduced social behavior, and hyperlocomotion, which are corrected by the atypical antipsychotic drug olanzapine. Additionally, synapsin II knock-down disrupts serial search efficiency. These behavioral changes are accompanied by reductions in vesicular neurotransmitter transporter protein concentrations for glutamate (VGLUT1 and VGLUT2) and GABA (VGAT), without affecting dopamine (VMAT2). These results implicate a causal role for decreased synapsin II in the medial prefrontal cortex in the pathophysiology of schizophrenia and the mechanisms of aberrant prefrontal cortical circuitry, and suggest that synapsin II may potentially serve as a novel therapeutic target for this disorder.
Collapse
Affiliation(s)
- Bailey A Dyck
- Department of Psychiatry and Behavioral Neurosciences, McMaster University, Hamilton, ON, Canada, L8N 3Z5
| | | | | | | |
Collapse
|
20
|
Fassio A, Raimondi A, Lignani G, Benfenati F, Baldelli P. Synapsins: from synapse to network hyperexcitability and epilepsy. Semin Cell Dev Biol 2011; 22:408-15. [PMID: 21816229 DOI: 10.1016/j.semcdb.2011.07.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Accepted: 07/13/2011] [Indexed: 01/31/2023]
Abstract
The synapsin family in mammals consists of at least 10 isoforms encoded by three distinct genes and composed by a mosaic of conserved and variable domains. Synapsins, although not essential for the basic development and functioning of neuronal networks, are extremely important for the fine-tuning of SV cycling and neuronal plasticity. Single, double and triple synapsin knockout mice, with the notable exception of the synapsin III knockout mice, show a severe epileptic phenotype without gross alterations in brain morphology and connectivity. However, the molecular and physiological mechanisms underlying the pathogenesis of the epileptic phenotype observed in synapsin deficient mice are still far from being elucidated. In this review, we summarize the current knowledge about the role of synapsins in the regulation of network excitability and about the molecular mechanism leading to epileptic phenotype in mouse lines lacking one or more synapsin isoforms. The current evidences indicate that synapsins exert distinct roles in excitatory versus inhibitory synapses by differentially affecting crucial steps of presynaptic physiology and by this mean participate in the determination of network hyperexcitability.
Collapse
Affiliation(s)
- Anna Fassio
- Department of Experimental Medicine, Section of Physiology and National Institute of Neuroscience, University of Genova, Genova, Italy
| | | | | | | | | |
Collapse
|
21
|
Bogen IL, Jensen V, Hvalby Ø, Walaas SI. Glutamatergic neurotransmission in the synapsin I and II double knock-out mouse. Semin Cell Dev Biol 2011; 22:400-7. [DOI: 10.1016/j.semcdb.2011.07.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Accepted: 07/13/2011] [Indexed: 01/19/2023]
|
22
|
Compensatory network alterations upon onset of epilepsy in synapsin triple knock-out mice. Neuroscience 2011; 189:108-22. [PMID: 21621590 DOI: 10.1016/j.neuroscience.2011.05.030] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Revised: 04/16/2011] [Accepted: 05/11/2011] [Indexed: 11/21/2022]
Abstract
Adult synapsin triple-knockout mice exhibit epilepsy that manifests as generalized tonic-clonic seizures. Because in vitro recordings have shown a reduction in quantal release from inhibitory neurons, an inherent excitation-inhibition imbalance has been hypothesized as the direct culprit for epilepsy in these mice. We critically assessed this hypothesis by examining neurotransmission during the emergence of epilepsy. Using long-term video and telemetric EEG monitoring we found that synapsin triple-knockout mice exhibit an abrupt transition during early adulthood from a seizure-free presymptomatic latent state to a consistent symptomatic state of sensory-induced seizures. Electrophysiological recordings showed that during the latent period larger field responses could be elicited in slices from mutant mice. However, only after the transition to a symptomatic state in the adult mice did evoked epileptiform activity become prevalent. This state was characterized by resistance to the epileptiform-promoting effects of 4-aminopyridine, by marked hypersensitivity to blockage of GABAA receptors, and by the emergence of unresponsiveness to NMDA receptor antagonism, all of which were not observed during the latent period. Importantly, enhancement in inhibitory transmission was associated with upregulation of GAD67 expression without affecting the number of inhibitory neurons in the same brain areas where epileptiform activity was recorded. We therefore suggest that while deletion of the synapsins initially increases cortical network activity, this enhanced excitability is insufficient to elicit seizures. Rather, compensatory epileptogenic mechanisms are activated during the latent period that lead to an additional almost-balanced enhancement of both the excitatory and inhibitory components of the network, finally culminating in the emergence of epilepsy.
Collapse
|
23
|
Diester I, Kaufman MT, Mogri M, Pashaie R, Goo W, Yizhar O, Ramakrishnan C, Deisseroth K, Shenoy KV. An optogenetic toolbox designed for primates. Nat Neurosci 2011; 14:387-97. [PMID: 21278729 DOI: 10.1038/nn.2749] [Citation(s) in RCA: 311] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 12/29/2010] [Indexed: 11/09/2022]
Abstract
Optogenetics is a technique for controlling subpopulations of neurons in the intact brain using light. This technique has the potential to enhance basic systems neuroscience research and to inform the mechanisms and treatment of brain injury and disease. Before launching large-scale primate studies, the method needs to be further characterized and adapted for use in the primate brain. We assessed the safety and efficiency of two viral vector systems (lentivirus and adeno-associated virus), two human promoters (human synapsin (hSyn) and human thymocyte-1 (hThy-1)) and three excitatory and inhibitory mammalian codon-optimized opsins (channelrhodopsin-2, enhanced Natronomonas pharaonis halorhodopsin and the step-function opsin), which we characterized electrophysiologically, histologically and behaviorally in rhesus monkeys (Macaca mulatta). We also introduced a new device for measuring in vivo fluorescence over time, allowing minimally invasive assessment of construct expression in the intact brain. We present a set of optogenetic tools designed for optogenetic experiments in the non-human primate brain.
Collapse
Affiliation(s)
- Ilka Diester
- Department of Bioengineering, Stanford University, Stanford, California, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
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
|