1
|
Xiao N, Yin L, Teopiz KM, Kwan ATH, Le GH, Wong S, Valentino K, Choi H, Rosenblat JD, Ho R, Lee S, McIntyre RS. The sigma-1 receptor: a mechanistically-informed therapeutic target for antidepressants. Expert Opin Ther Targets 2025:1-15. [PMID: 40298911 DOI: 10.1080/14728222.2025.2500424] [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: 12/02/2024] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
INTRODUCTION The mechanism of action of antidepressants is not fully ascertained. In addition to monoamines, disparate other effectors are also implicated in the molecular and cellular effects of chronic stress including neurogenesis, neurodifferentiation, and neuroplasticity. Evidence suggests sigma-1 receptors (S1Rs) as a putative target and possible mediator of antidepressant activity. AREAS COVERED Data from preclinical and clinical trials was synthesized from inception to August 2024. Results showed that S1Rs regulate neurotransmitter availability and release (e.g. monoamines, glutamate), and influence intracellular Ca2+ levels as well as immune inflammatory responses. The introduction of the N-Methyl-D-aspartic Acid (NMDA) antagonist/S1R agonist dextromethorphan-bupropion in August of 2022 represented the first time the Food and Drug Administration (FDA) permitted language that the hypothesized mechanism of an antidepressant involved activity at S1Rs. We also describe the physiology, pathophysiology, and function of S1Rs. EXPERT OPINION Sigma-1 modulation is relevant to the mechanism of action of agents currently FDA-approved in major depressive disorder (MDD) (e.g. dextromethorphan-bupropion). Modulating sigma-1 systems is fit for purpose as it relates to future therapeutic discoveries and development in depressive and other mental disorders. Whether sigma-1 modulation is uniquely relevant to targeting dimensions of psychopathology that are more difficult to treat (i.e. anhedonia) awaits determination.
Collapse
Affiliation(s)
- Naomi Xiao
- Department of Research, Brain and Cognition Discovery Foundation, Toronto, Canada
- Department of Health Sciences, Queen's University, Kingston, Canada
| | - Liyang Yin
- Department of Research, Brain and Cognition Discovery Foundation, Toronto, Canada
| | - Kayla M Teopiz
- Department of Research, Brain and Cognition Discovery Foundation, Toronto, Canada
| | - Angela T H Kwan
- Department of Research, Brain and Cognition Discovery Foundation, Toronto, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Gia Han Le
- Department of Research, Brain and Cognition Discovery Foundation, Toronto, Canada
- Mood Disorder Psychopharmacology Unit, University Health Network, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Sabrina Wong
- Department of Research, Brain and Cognition Discovery Foundation, Toronto, Canada
- Mood Disorder Psychopharmacology Unit, University Health Network, Toronto, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Kyle Valentino
- Department of Research, Brain and Cognition Discovery Foundation, Toronto, Canada
- Mood Disorder Psychopharmacology Unit, University Health Network, Toronto, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Hayun Choi
- Department of Research, Brain and Cognition Discovery Foundation, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
- Department of Psychiatry, Veteran Health Service Medical Center, Seoul, Republic of Korea
| | - Joshua D Rosenblat
- Mood Disorder Psychopharmacology Unit, University Health Network, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Roger Ho
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore
- Division of Life Science (LIFS), Hong Kong University of Science and Technology (HKUST), Clear Water Bay, Hong Kong
| | - Serene Lee
- Department of Research, Brain and Cognition Discovery Foundation, Toronto, Canada
- Department of Health Sciences, Queen's University, Kingston, Canada
| | - Roger S McIntyre
- Department of Psychiatry, University of Toronto, Toronto, Canada
| |
Collapse
|
2
|
Zhang C, Zhou T, Qiao S, Lu L, Zhu M, Wang A, Zhang S. Taurine Attenuates Neuronal Ferroptosis by CSF-Derived Exosomes of GABABR Encephalitis Through GABABR/NF2/P-YAP Pathway. Mol Neurobiol 2025:10.1007/s12035-025-04819-3. [PMID: 40085353 DOI: 10.1007/s12035-025-04819-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
GABAB receptor (GABABR) encephalitis represents a rare subtype of paraneoplastic limbic encephalitis (LE), characterized by persistent seizures and cognitive impairments. Nevertheless, the precise phenotype and underlying mechanisms of neuronal dysfunction associated with intrathecal lymphocytes in GABABR encephalitis remain inadequately understood. In the present study, we demonstrate that exosomes derived from the cerebrospinal fluid (CSF) of patients with GABABR encephalitis can induce neuronal ferroptosis, oxidative stress, iron accumulation, and lipid hyperoxidation in an in vitro model of anti-GABABR encephalitis. MicroRNA (miRNA) sequencing revealed that miR-92a-3p is a differentially expressed miRNA in CSF exosomes, and its expression was positively correlated with unfavorable clinical outcomes in GABABR encephalitis patients during a 6-month follow-up period. The NF2/P-YAP signaling pathway was identified as a downstream effector of miR-92a-3p, influencing the expression of ACSL4/GPX4 and IL-6, with the expression of these genes being enhanced following taurine supplementation. Clinically, taurine levels in CSF exhibited a negative correlation with IL-6 levels, CSF cell counts, blood-CSF barrier integrity, and clinical prognosis in GABABR encephalitis. Mechanistically, taurine effectively reduced reactive oxygen species (ROS) and iron accumulation, as well as IL-6 production, while modulating the levels of NF2, P-YAP, ACSL4, and GPX4 in neurons treated with CSF-derived exosomes from GABABR encephalitis through GABABR activation. Proliferation assays indicated that extracellular taurine intake activated CD4 + T cells, CD8 + T cells, and CD19 + B cells in the CSF of patients with GABABR encephalitis. In summary, our findings reveal for the first time that intrathecal lymphocytes in GABABR encephalitis maintain an activated state by absorbing extracellular taurine and that decreased taurine levels in CSF promote neuronal ferroptosis via the miR-92a-3p-mediated NF2/P-YAP/ACSL4 pathway.
Collapse
Affiliation(s)
- Chong Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Shandong Institute of Neuroimmunology, Jinan, China
- Shandong First Medical University, Jinan, China
| | - Tianyu Zhou
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Shandong Institute of Neuroimmunology, Jinan, China
- Shandong First Medical University, Jinan, China
| | - Shan Qiao
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Shandong Institute of Neuroimmunology, Jinan, China
| | - Lu Lu
- Department of Neurology, Linyi People's Hospital, Linyi, China
| | - Meirong Zhu
- Department of Critical Care Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Aihua Wang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Shandong Institute of Neuroimmunology, Jinan, China
| | - Shanchao Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Shandong Institute of Neuroimmunology, Jinan, China.
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Ren X, Sirois CL, Doudlah R, Mendez-Albelo NM, Hai A, Rosenberg A, Zhao X. A Semi-Automated MEA Spike sorting (SAMS) method for high throughput assessment of cultured neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.08.637245. [PMID: 39975344 PMCID: PMC11839033 DOI: 10.1101/2025.02.08.637245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Neurons derived from human pluripotent stem cells (hPSCs) are valuable models for studying brain development and developing therapies for brain disorders. Evaluating human-derived neurons requires assessing their electrical activity, which can be achieved using multi-electrode arrays (MEAs) for extracellular recordings. Because each electrode channel generally detects activity from multiple neurons, resolving the activity of single neurons requires a process called spike sorting. However, currently available spike sorting methods are not optimized for the analysis of hPSC-derived neurons, and require complex workflows and time-consuming manual intervention. Here, we introduce a S emi- A utomated M EA S pike sorting software (SAMS) designed specifically for low-density MEA recordings of cultured neurons. SAMS outperforms commercially available automated spike sorting algorithms in terms of accuracy and greatly reduces computational and human processing time. By providing an accessible, efficient, and integrated platform for spike sorting, SAMS enhances the resolution and utility of MEA in disease modeling and drug development using human-derived neurons. Highlights SAMS is designed and optimized for high throughput analysis of hPSC-derived neurons.SAMS is more efficient and accurate compared to recommended spike-sorting software.SAMS resolves phenotypic differences previously not observed without spike sorting.SAMS is an open-source software.
Collapse
|
4
|
Cai Y, Wang T. Regulation of presynaptic homeostatic plasticity by glial signalling in Alzheimer's disease. J Physiol 2024. [PMID: 39705214 DOI: 10.1113/jp286751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 12/04/2024] [Indexed: 12/22/2024] Open
Abstract
Alzheimer's disease (AD), the most common form of dementia among the elderly, affects numerous individuals worldwide. Despite advances in understanding the molecular underpinnings of AD pathology, effective treatments to prevent or cure the disease remain elusive. AD is characterized not only by pathological hallmarks such as amyloid plaques and neurofibrillary tangles but also by impairments in synaptic physiology, circuit activity and cognitive function. Synaptic homeostatic plasticity plays a vital role in maintaining the stability of synaptic and neural functions amid genetic and environmental disturbances. A key component of this regulation is presynaptic homeostatic potentiation, where increased presynaptic neurotransmitter release compensates for reduced postsynaptic glutamate receptor functionality, thereby stabilizing neuronal excitability. The role of presynaptic homeostatic plasticity in synapse stabilization in AD, however, remains unclear. Moreover, recent advances in transcriptomics have illuminated the complex roles of glial cells in regulating synaptic function in ageing brains and in the progression of neurodegenerative diseases. Yet, the impact of AD-related abnormalities in glial signalling on synaptic homeostatic plasticity has not been fully delineated. This review discusses recent findings on how glial dysregulation in AD affects presynaptic homeostatic plasticity. There is increasing evidence that disrupted glial signalling, particularly through aberrant histone acetylation and transcriptomic changes in glia, compromises this plasticity in AD. Notably, the sphingosine signalling pathway has been identified as being protective in stabilizing synaptic physiology through epigenetic and homeostatic mechanisms, presenting potential therapeutic targets for treating neurodegenerative disorders.
Collapse
Affiliation(s)
- Yimei Cai
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, D.C., USA
| | - Tingting Wang
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, D.C., USA
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, D.C., USA
| |
Collapse
|
5
|
Gao S, Shen R, Li J, Jiang Y, Sun H, Wu X, Li X, Miao C, He M, Wang J, Chen W. N-acetyltransferase 10 mediates cognitive dysfunction through the acetylation of GABA BR1 mRNA in sepsis-associated encephalopathy. Proc Natl Acad Sci U S A 2024; 121:e2410564121. [PMID: 39190359 PMCID: PMC11388286 DOI: 10.1073/pnas.2410564121] [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: 06/01/2024] [Accepted: 07/30/2024] [Indexed: 08/28/2024] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a critical neurological complication of sepsis and represents a crucial factor contributing to high mortality and adverse prognosis in septic patients. This study explored the contribution of NAT10-mediated messenger RNA (mRNA) acetylation in cognitive dysfunction associated with SAE, utilizing a cecal ligation and puncture (CLP)-induced SAE mouse model. Our findings demonstrate that CLP significantly upregulates NAT10 expression and mRNA acetylation in the excitatory neurons of the hippocampal dentate gyrus (DG). Notably, neuronal-specific Nat10 knockdown improved cognitive function in septic mice, highlighting its critical role in SAE. Proteomic analysis, RNA immunoprecipitation, and real-time qPCR identified GABABR1 as a key downstream target of NAT10. Nat10 deletion reduced GABABR1 expression, and subsequently weakened inhibitory postsynaptic currents in hippocampal DG neurons. Further analysis revealed that microglia activation and the release of inflammatory mediators lead to the increased NAT10 expression in neurons. Microglia depletion with PLX3397 effectively reduced NAT10 and GABABR1 expression in neurons, and ameliorated cognitive dysfunction induced by SAE. In summary, our findings revealed that after CLP, NAT10 in hippocampal DG neurons promotes GABABR1 expression through mRNA acetylation, leading to cognitive dysfunction.
Collapse
Affiliation(s)
- Shenjia Gao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai200032, China
| | - Ruling Shen
- Shanghai Laboratory Animal Research Center, Shanghai201203, China
| | - Jie Li
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education (MOE) Frontiers Center for Brain Science, Shanghai200032, China
- Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai200032, China
| | - Yi Jiang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai200032, China
| | - Hao Sun
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai200032, China
| | - Xinyi Wu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai200032, China
| | - Xiya Li
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai200032, China
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai200032, China
| | - Miao He
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education (MOE) Frontiers Center for Brain Science, Shanghai200032, China
- Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai200032, China
| | - Jun Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai200032, China
| | - Wankun Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai200032, China
- Department of Anesthesiology, Shanghai Geriatric Medical Center, Shanghai201104, China
- Department of Anesthesiology, QingPu Branch of Zhongshan Hospital, Fudan University, Shanghai201799, China
| |
Collapse
|
6
|
Kundu S, Paul B, Reuevni I, Lamprecht R, Barkai E. Learning-induced bidirectional enhancement of inhibitory synaptic metaplasticity. J Physiol 2024; 602:2343-2358. [PMID: 38654583 DOI: 10.1113/jp284761] [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: 03/28/2023] [Accepted: 04/02/2024] [Indexed: 04/26/2024] Open
Abstract
Training rodents in a particularly difficult olfactory-discrimination (OD) task results in the acquisition of the ability to perform the task well, termed 'rule learning'. In addition to enhanced intrinsic excitability and synaptic excitation in piriform cortex pyramidal neurons, rule learning results in increased synaptic inhibition across the whole cortical network to the point where it precisely maintains the balance between inhibition and excitation. The mechanism underlying such precise inhibitory enhancement remains to be explored. Here, we use brain slices from transgenic mice (VGAT-ChR2-EYFP), enabling optogenetic stimulation of single GABAergic neurons and recordings of unitary synaptic events in pyramidal neurons. Quantal analysis revealed that learning-induced enhanced inhibition is mediated by increased quantal size of the evoked inhibitory events. Next, we examined the plasticity of synaptic inhibition induced by long-lasting, intrinsically evoked spike firing in post-synaptic neurons. Repetitive depolarizing current pulses from depolarized (-70 mV) or hyperpolarized (-90 mV) membrane potentials induced long-term depression (LTD) and long-term potentiation (LTP) of synaptic inhibition, respectively. We found a profound bidirectional increase in the ability to induce both LTD, mediated by L-type calcium channels, and LTP, mediated by R-type calcium channels after rule learning. Blocking the GABAB receptor reversed the effect of intrinsic stimulation at -90 mV from LTP to LTD. We suggest that learning greatly enhances the ability to modify the strength of synaptic inhibition of principal neurons in both directions. Such plasticity of synaptic plasticity allows fine-tuning of inhibition on each particular neuron, thereby stabilizing the network while maintaining the memory of the rule. KEY POINTS: Olfactory discrimination rule learning results in long-lasting enhancement of synaptic inhibition on piriform cortex pyramidal neurons. Quantal analysis of unitary inhibitory synaptic events, evoked by optogenetic minimal stimulation, revealed that enhanced synaptic inhibition is mediated by increased quantal size. Surprisingly, metaplasticity of synaptic inhibition, induced by intrinsically evoked repetitive spike firing, is increased bidirectionally. The susceptibility to both long-term depression (LTD) and long-term potentiation (LTP) of inhibition is enhanced after learning. LTD of synaptic inhibition is mediated by L-type calcium channels and LTP by R-type calcium channels. LTP is also dependent on activation of GABAB receptors. We suggest that learning-induced changes in the metaplasticity of synaptic inhibition enable the fine-tuning of inhibition on each particular neuron, thereby stabilizing the network while maintaining the memory of the rule.
Collapse
Affiliation(s)
- Sankhanava Kundu
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Blesson Paul
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Iris Reuevni
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Raphael Lamprecht
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Edi Barkai
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
7
|
Slutsky I. Linking activity dyshomeostasis and sleep disturbances in Alzheimer disease. Nat Rev Neurosci 2024; 25:272-284. [PMID: 38374463 DOI: 10.1038/s41583-024-00797-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2024] [Indexed: 02/21/2024]
Abstract
The presymptomatic phase of Alzheimer disease (AD) starts with the deposition of amyloid-β in the cortex and begins a decade or more before the emergence of cognitive decline. The trajectory towards dementia and neurodegeneration is shaped by the pathological load and the resilience of neural circuits to the effects of this pathology. In this Perspective, I focus on recent advances that have uncovered the vulnerability of neural circuits at early stages of AD to hyperexcitability, particularly when the brain is in a low-arousal states (such as sleep and anaesthesia). Notably, this hyperexcitability manifests before overt symptoms such as sleep and memory deficits. Using the principles of control theory, I analyse the bidirectional relationship between homeostasis of neuronal activity and sleep and propose that impaired activity homeostasis during sleep leads to hyperexcitability and subsequent sleep disturbances, whereas sleep disturbances mitigate hyperexcitability via negative feedback. Understanding the interplay among activity homeostasis, neuronal excitability and sleep is crucial for elucidating the mechanisms of vulnerability to and resilience against AD pathology and for identifying new therapeutic avenues.
Collapse
Affiliation(s)
- Inna Slutsky
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
8
|
Benke D, Bhat MA, Hleihil M. GABAB Receptors: Molecular Organization, Function, and Alternative Drug Development by Targeting Protein-Protein Interactions. THE RECEPTORS 2024:3-39. [DOI: 10.1007/978-3-031-67148-7_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
9
|
Garrudo FFF, Linhardt RJ, Ferreira FC, Morgado J. Designing Electrical Stimulation Platforms for Neural Cell Cultivation Using Poly(aniline): Camphorsulfonic Acid. Polymers (Basel) 2023; 15:2674. [PMID: 37376320 DOI: 10.3390/polym15122674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/01/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Electrical stimulation is a powerful strategy to improve the differentiation of neural stem cells into neurons. Such an approach can be implemented, in association with biomaterials and nanotechnology, for the development of new therapies for neurological diseases, including direct cell transplantation and the development of platforms for drug screening and disease progression evaluation. Poly(aniline):camphorsulfonic acid (PANI:CSA) is one of the most well-studied electroconductive polymers, capable of directing an externally applied electrical field to neural cells in culture. There are several examples in the literature on the development of PANI:CSA-based scaffolds and platforms for electrical stimulation, but no review has examined the fundamentals and physico-chemical determinants of PANI:CSA for the design of platforms for electrical stimulation. This review evaluates the current literature regarding the application of electrical stimulation to neural cells, specifically reviewing: (1) the fundamentals of bioelectricity and electrical stimulation; (2) the use of PANI:CSA-based systems for electrical stimulation of cell cultures; and (3) the development of scaffolds and setups to support the electrical stimulation of cells. Throughout this work, we critically evaluate the revised literature and provide a steppingstone for the clinical application of the electrical stimulation of cells using electroconductive PANI:CSA platforms/scaffolds.
Collapse
Affiliation(s)
- Fábio F F Garrudo
- Instituto de Telecomunicações, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001 Lisbon, Portugal
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001 Lisbon, Portugal
- iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Avenida Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Robert J Linhardt
- Department of Chemical and Biological Engineering, Biology and Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA
| | - Frederico Castelo Ferreira
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001 Lisbon, Portugal
- iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Avenida Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Jorge Morgado
- Instituto de Telecomunicações, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001 Lisbon, Portugal
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001 Lisbon, Portugal
| |
Collapse
|
10
|
Bar L, Shalom L, Lezmy J, Peretz A, Attali B. Excitatory and inhibitory hippocampal neurons differ in their homeostatic adaptation to chronic M-channel modulation. Front Mol Neurosci 2022; 15:972023. [PMID: 36311018 PMCID: PMC9614320 DOI: 10.3389/fnmol.2022.972023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/27/2022] [Indexed: 12/03/2022] Open
Abstract
A large body of studies has investigated bidirectional homeostatic plasticity both in vitro and in vivo using numerous pharmacological manipulations of activity or behavioral paradigms. However, these experiments rarely explored in the same cellular system the bidirectionality of the plasticity and simultaneously on excitatory and inhibitory neurons. M-channels are voltage-gated potassium channels that play a crucial role in regulating neuronal excitability and plasticity. In cultured hippocampal excitatory neurons, we previously showed that chronic exposure to the M-channel blocker XE991 leads to adaptative compensations, thereby triggering at different timescales intrinsic and synaptic homeostatic plasticity. This plastic adaptation barely occurs in hippocampal inhibitory neurons. In this study, we examined whether this homeostatic plasticity induced by M-channel inhibition was bidirectional by investigating the acute and chronic effects of the M-channel opener retigabine on hippocampal neuronal excitability. Acute retigabine exposure decreased excitability in both excitatory and inhibitory neurons. Chronic retigabine treatment triggered in excitatory neurons homeostatic adaptation of the threshold current and spontaneous firing rate at a time scale of 4–24 h. These plastic changes were accompanied by a substantial decrease in the M-current density and by a small, though significant, proximal relocation of Kv7.3-FGF14 segment along the axon initial segment. Thus, bidirectional homeostatic changes were observed in excitatory neurons though not symmetric in kinetics and mechanisms. Contrastingly, in inhibitory neurons, the compensatory changes in intrinsic excitability barely occurred after 48 h, while no homeostatic normalization of the spontaneous firing rate was observed. Our results indicate that excitatory and inhibitory hippocampal neurons differ in their adaptation to chronic alterations in neuronal excitability induced by M-channel bidirectional modulation.
Collapse
|
11
|
Unravelling biological roles and mechanisms of GABA BR on addiction and depression through mood and memory disorders. Biomed Pharmacother 2022; 155:113700. [PMID: 36152411 DOI: 10.1016/j.biopha.2022.113700] [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: 08/12/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/23/2022] Open
Abstract
The metabotropic γ-aminobutyric acid type B receptor (GABABR) remains a hotspot in the recent research area. Being an idiosyncratic G-protein coupled receptor family member, the GABABR manifests adaptively tailored functionality under multifarious modulations by a constellation of agents, pointing to cross-talk between receptors and effectors that converge on the domains of mood and memory. This review systematically summarizes the latest achievements in signal transduction mechanisms of the GABABR-effector-regulator complex and probes how the up-and down-regulation of membrane-delimited GABABRs are associated with manifold intrinsic and extrinsic agents in synaptic strength and plasticity. Neuropsychiatric conditions depression and addiction share the similar pathophysiology of synapse inadaptability underlying negative mood-related processes, memory formations, and impairments. In the attempt to emphasize all convergent discoveries, we hope the insights gained on the GABABR system mechanisms of action are conducive to designing more therapeutic candidates so as to refine the prognosis rate of diseases and minimize side effects.
Collapse
|
12
|
IGF-1 receptor regulates upward firing rate homeostasis via the mitochondrial calcium uniporter. Proc Natl Acad Sci U S A 2022; 119:e2121040119. [PMID: 35943986 PMCID: PMC9388073 DOI: 10.1073/pnas.2121040119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
An emerging hypothesis is that neuronal circuits homeostatically maintain a stable spike rate despite continuous environmental changes. This firing rate homeostasis is believed to confer resilience to neurodegeneration and cognitive decline. We show that insulin-like growth factor-1 receptor (IGF-1R) is necessary for homeostatic response of mean firing rate to inactivity, termed “upward firing rate homeostasis.” We show that its mechanism of action is to couple spike bursts with downstream mitochondrial Ca2+ influx via the mitochondrial calcium uniporter complex (MCUc). We propose that MCUc is a homeostatic Ca2+ sensor that triggers the integrated homeostatic response. Firing rate homeostasis may be the principal mechanism by which IGF-1R regulates aging and neurodevelopmental and neurodegenerative disorders. Regulation of firing rate homeostasis constitutes a fundamental property of central neural circuits. While intracellular Ca2+ has long been hypothesized to be a feedback control signal, the molecular machinery enabling a network-wide homeostatic response remains largely unknown. We show that deletion of insulin-like growth factor-1 receptor (IGF-1R) limits firing rate homeostasis in response to inactivity, without altering the distribution of baseline firing rates. The deficient firing rate homeostatic response was due to disruption of both postsynaptic and intrinsic plasticity. At the cellular level, we detected a fraction of IGF-1Rs in mitochondria, colocalized with the mitochondrial calcium uniporter complex (MCUc). IGF-1R deletion suppressed transcription of the MCUc members and burst-evoked mitochondrial Ca2+ (mitoCa2+) by weakening mitochondria-to-cytosol Ca2+ coupling. Overexpression of either mitochondria-targeted IGF-1R or MCUc in IGF-1R–deficient neurons was sufficient to rescue the deficits in burst-to-mitoCa2+ coupling and firing rate homeostasis. Our findings indicate that mitochondrial IGF-1R is a key regulator of the integrated homeostatic response by tuning the reliability of burst transfer by MCUc. Based on these results, we propose that MCUc acts as a homeostatic Ca2+ sensor. Faulty activation of MCUc may drive dysregulation of firing rate homeostasis in aging and in brain disorders associated with aberrant IGF-1R/MCUc signaling.
Collapse
|
13
|
Avoli M, Lévesque M. GABA B Receptors: are they Missing in Action in Focal Epilepsy Research? Curr Neuropharmacol 2022; 20:1704-1716. [PMID: 34429053 PMCID: PMC9881065 DOI: 10.2174/1570159x19666210823102332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/24/2021] [Accepted: 08/07/2021] [Indexed: 11/22/2022] Open
Abstract
GABA, the key inhibitory neurotransmitter in the adult forebrain, activates pre- and postsynaptic receptors that have been categorized as GABAA, which directly open ligand-gated (or receptor-operated) ion-channels, and GABAB, which are metabotropic since they operate through second messengers. Over the last three decades, several studies have addressed the role of GABAB receptors in the pathophysiology of generalized and focal epileptic disorders. Here, we will address their involvement in focal epileptic disorders by mainly reviewing in vitro studies that have shown: (i) how either enhancing or decreasing GABAB receptor function can favour epileptiform synchronization and thus ictogenesis, although with different features; (ii) the surprising ability of GABAB receptor antagonism to disclose ictal-like activity when the excitatory ionotropic transmission is abolished; and (iii) their contribution to controlling seizure-like discharges during repetitive electrical stimuli delivered in limbic structures. In spite of this evidence, the role of GABAB receptor function in focal epileptic disorders has been attracting less interest when compared to the numerous studies that have addressed GABAA receptor signaling. Therefore, the main aim of our mini-review is to revive interest in the function of GABAB receptors in focal epilepsy research.
Collapse
Affiliation(s)
- Massimo Avoli
- Montreal Neurological Institute-Hospital and Departments of Neurology & Neurosurgery and of; ,Department of Experimental Medicine, Sapienza University of Rome, 00185Rome, Italy,Address correspondence to this author at the Montreal Neurological Institute-Hospital, 3801 University Street, Montréal, Canada, H3A 2B4, QC; Tels: +1 514 998 6790; +39 333 483 1060; E-mail:
| | - Maxime Lévesque
- Montreal Neurological Institute-Hospital and Departments of Neurology & Neurosurgery and of;
| |
Collapse
|
14
|
Keeping the Balance: GABAB Receptors in the Developing Brain and Beyond. Brain Sci 2022; 12:brainsci12040419. [PMID: 35447949 PMCID: PMC9031223 DOI: 10.3390/brainsci12040419] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 12/16/2022] Open
Abstract
The main neurotransmitter in the brain responsible for the inhibition of neuronal activity is γ-aminobutyric acid (GABA). It plays a crucial role in circuit formation during development, both via its primary effects as a neurotransmitter and also as a trophic factor. The GABAB receptors (GABABRs) are G protein-coupled metabotropic receptors; on one hand, they can influence proliferation and migration; and, on the other, they can inhibit cells by modulating the function of K+ and Ca2+ channels, doing so on a slower time scale and with a longer-lasting effect compared to ionotropic GABAA receptors. GABABRs are expressed pre- and post-synaptically, at both glutamatergic and GABAergic terminals, thus being able to shape neuronal activity, plasticity, and the balance between excitatory and inhibitory synaptic transmission in response to varying levels of extracellular GABA concentration. Furthermore, given their subunit composition and their ability to form complexes with several associated proteins, GABABRs display heterogeneity with regard to their function, which makes them a promising target for pharmacological interventions. This review will describe (i) the latest results concerning GABABRs/GABABR-complex structures, their function, and the developmental time course of their appearance and functional integration in the brain, (ii) their involvement in manifestation of various pathophysiological conditions, and (iii) the current status of preclinical and clinical studies involving GABABR-targeting drugs.
Collapse
|
15
|
Zarhin D, Atsmon R, Ruggiero A, Baeloha H, Shoob S, Scharf O, Heim LR, Buchbinder N, Shinikamin O, Shapira I, Styr B, Braun G, Harel M, Sheinin A, Geva N, Sela Y, Saito T, Saido T, Geiger T, Nir Y, Ziv Y, Slutsky I. Disrupted neural correlates of anesthesia and sleep reveal early circuit dysfunctions in Alzheimer models. Cell Rep 2022; 38:110268. [PMID: 35045289 PMCID: PMC8789564 DOI: 10.1016/j.celrep.2021.110268] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/06/2021] [Accepted: 12/22/2021] [Indexed: 11/30/2022] Open
Abstract
Dysregulated homeostasis of neural activity has been hypothesized to drive Alzheimer's disease (AD) pathogenesis. AD begins with a decades-long presymptomatic phase, but whether homeostatic mechanisms already begin failing during this silent phase is unknown. We show that before the onset of memory decline and sleep disturbances, familial AD (fAD) model mice display no deficits in CA1 mean firing rate (MFR) during active wakefulness. However, homeostatic down-regulation of CA1 MFR is disrupted during non-rapid eye movement (NREM) sleep and general anesthesia in fAD mouse models. The resultant hyperexcitability is attenuated by the mitochondrial dihydroorotate dehydrogenase (DHODH) enzyme inhibitor, which tunes MFR toward lower set-point values. Ex vivo fAD mutations impair downward MFR homeostasis, resulting in pathological MFR set points in response to anesthetic drug and inhibition blockade. Thus, firing rate dyshomeostasis of hippocampal circuits is masked during active wakefulness but surfaces during low-arousal brain states, representing an early failure of the silent disease stage.
Collapse
Affiliation(s)
- Daniel Zarhin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Refaela Atsmon
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Antonella Ruggiero
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Halit Baeloha
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Shiri Shoob
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Oded Scharf
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Leore R Heim
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Nadav Buchbinder
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ortal Shinikamin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ilana Shapira
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Boaz Styr
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Gabriella Braun
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Michal Harel
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Anton Sheinin
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Nitzan Geva
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yaniv Sela
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama 351-0198, Japan; Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Takaomi Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Tamar Geiger
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yuval Nir
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yaniv Ziv
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
16
|
Barak-Broner N, Singer-Lahat D, Chikvashvili D, Lotan I. CK2 Phosphorylation Is Required for Regulation of Syntaxin 1A Activity in Ca 2+-Triggered Release in Neuroendocrine Cells. Int J Mol Sci 2021; 22:ijms222413556. [PMID: 34948351 PMCID: PMC8708312 DOI: 10.3390/ijms222413556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 12/29/2022] Open
Abstract
The polybasic juxtamembrane region (5RK) of the plasma membrane neuronal SNARE, syntaxin1A (Syx), was previously shown by us to act as a fusion clamp in PC12 cells, as charge neutralization of 5RK promotes spontaneous and inhibits Ca2+-triggered release. Using a Syx-based FRET probe (CSYS), we demonstrated that 5RK is required for a depolarization-induced Ca+2-dependent opening (close-to-open transition; CDO) of Syx, which involves the vesicular SNARE synaptobrevin2 and occurs concomitantly with Ca2+-triggered release. Here, we investigated the mechanism underlying the CDO requirement for 5RK and identified phosphorylation of Syx at Ser-14 (S14) by casein kinase 2 (CK2) as a crucial molecular determinant. Thus, following biochemical verification that both endogenous Syx and CSYS are constitutively S14 phosphorylated in PC12 cells, dynamic FRET analysis of phospho-null and phospho-mimetic mutants of CSYS and the use of a CK2 inhibitor revealed that the S14 phosphorylation confers the CDO requirement for 5RK. In accord, amperometric analysis of catecholamine release revealed that the phospho-null mutant does not support Ca2+-triggered release. These results identify a functionally important CK2 phosphorylation of Syx that is required for the 5RK-regulation of CDO and for concomitant Ca2+-triggered release. Further, also spontaneous release, conferred by charge neutralization of 5RK, was abolished in the phospho-null mutant.
Collapse
Affiliation(s)
- Noa Barak-Broner
- Department of Neurobiology Biochemistry & Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv-Yafo 69978, Israel;
| | - Dafna Singer-Lahat
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv-Yafo 69978, Israel; (D.S.-L.); (D.C.)
| | - Dodo Chikvashvili
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv-Yafo 69978, Israel; (D.S.-L.); (D.C.)
| | - Ilana Lotan
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv-Yafo 69978, Israel; (D.S.-L.); (D.C.)
- Sagol School of Neuroscience, Tel Aviv University, Ramat Aviv, Tel Aviv-Yafo 69978, Israel
- Correspondence:
| |
Collapse
|
17
|
Gustorff C, Scheuer T, Schmitz T, Bührer C, Endesfelder S. GABA B Receptor-Mediated Impairment of Intermediate Progenitor Maturation During Postnatal Hippocampal Neurogenesis of Newborn Rats. Front Cell Neurosci 2021; 15:651072. [PMID: 34421540 PMCID: PMC8377254 DOI: 10.3389/fncel.2021.651072] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 07/12/2021] [Indexed: 12/19/2022] Open
Abstract
The neurotransmitter GABA and its receptors assume essential functions during fetal and postnatal brain development. The last trimester of a human pregnancy and early postnatal life involves a vulnerable period of brain development. In the second half of gestation, there is a developmental shift from depolarizing to hyperpolarizing in the GABAergic system, which might be disturbed by preterm birth. Alterations of the postnatal GABA shift are associated with several neurodevelopmental disorders. In this in vivo study, we investigated neurogenesis in the dentate gyrus (DG) in response to daily administration of pharmacological GABAA (DMCM) and GABAB (CGP 35348) receptor inhibitors to newborn rats. Six-day-old Wistar rats (P6) were daily injected (i.p.) to postnatal day 11 (P11) with DMCM, CGP 35348, or vehicle to determine the effects of both antagonists on postnatal neurogenesis. Due to GABAB receptor blockade by CGP 35348, immunohistochemistry revealed a decrease in the number of NeuroD1 positive intermediate progenitor cells and a reduction of proliferative Nestin-positive neuronal stem cells at the DG. The impairment of hippocampal neurogenesis at this stage of differentiation is in line with a significantly decreased RNA expression of the transcription factors Pax6, Ascl1, and NeuroD1. Interestingly, the number of NeuN-positive postmitotic neurons was not affected by GABAB receptor blockade, although strictly associated transcription factors for postmitotic neurons, Tbr1, Prox1, and NeuroD2, displayed reduced expression levels, suggesting impairment by GABAB receptor antagonization at this stage of neurogenesis. Antagonization of GABAB receptors decreased the expression of neurotrophins (BDNF, NT-3, and NGF). In contrast to the GABAB receptor blockade, the GABAA receptor antagonization revealed no significant changes in cell counts, but an increased transcriptional expression of Tbr1 and Tbr2. We conclude that GABAergic signaling via the metabotropic GABAB receptor is crucial for hippocampal neurogenesis at the time of rapid brain growth and of the postnatal GABA shift. Differentiation and proliferation of intermediate progenitor cells are dependent on GABA. These insights become more pertinent in preterm infants whose developing brains are prematurely exposed to spostnatal stress and predisposed to poor neurodevelopmental disorders, possibly as sequelae of early disruption in GABAergic signaling.
Collapse
Affiliation(s)
- Charlotte Gustorff
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Till Scheuer
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas Schmitz
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Bührer
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|
18
|
Baker MR, Wong RY. Npas4a expression in the teleost forebrain is associated with stress coping style differences in fear learning. Sci Rep 2021; 11:12074. [PMID: 34103598 PMCID: PMC8187387 DOI: 10.1038/s41598-021-91495-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 05/21/2021] [Indexed: 12/14/2022] Open
Abstract
Learning to anticipate potentially dangerous contexts is an adaptive behavioral response to coping with stressors. An animal's stress coping style (e.g. proactive-reactive axis) is known to influence how it encodes salient events. However, the neural and molecular mechanisms underlying these stress coping style differences in learning are unknown. Further, while a number of neuroplasticity-related genes have been associated with alternative stress coping styles, it is unclear if these genes may bias the development of conditioned behavioral responses to stressful stimuli, and if so, which brain regions are involved. Here, we trained adult zebrafish to associate a naturally aversive olfactory cue with a given context. Next, we investigated if expression of two neural plasticity and neurotransmission-related genes (npas4a and gabbr1a) were associated with the contextual fear conditioning differences between proactive and reactive stress coping styles. Reactive zebrafish developed a stronger conditioned fear response and showed significantly higher npas4a expression in the medial and lateral zones of the dorsal telencephalon (Dm, Dl), and the supracommissural nucleus of the ventral telencephalon (Vs). Our findings suggest that the expression of activity-dependent genes like npas4a may be differentially expressed across several interconnected forebrain regions in response to fearful stimuli and promote biases in fear learning among different stress coping styles.
Collapse
Affiliation(s)
- Matthew R Baker
- Department of Biology, University of Nebraska at Omaha, Omaha, USA
| | - Ryan Y Wong
- Department of Biology, University of Nebraska at Omaha, Omaha, USA.
- Department of Psychology, University of Nebraska at Omaha, 6001 Dodge St, Omaha, NE, 68182, USA.
| |
Collapse
|
19
|
Ruggiero A, Katsenelson M, Slutsky I. Mitochondria: new players in homeostatic regulation of firing rate set points. Trends Neurosci 2021; 44:605-618. [PMID: 33865626 DOI: 10.1016/j.tins.2021.03.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/10/2021] [Accepted: 03/11/2021] [Indexed: 10/21/2022]
Abstract
Neural circuit functions are stabilized by homeostatic processes at long timescales in response to changes in behavioral states, experience, and learning. However, it remains unclear which specific physiological variables are being stabilized and which cellular or neural network components compose the homeostatic machinery. At this point, most evidence suggests that the distribution of firing rates among neurons in a neuronal circuit is the key variable that is maintained around a set-point value in a process called 'firing rate homeostasis.' Here, we review recent findings that implicate mitochondria as central players in mediating firing rate homeostasis. While mitochondria are known to regulate neuronal variables such as synaptic vesicle release or intracellular calcium concentration, the mitochondrial signaling pathways that are essential for firing rate homeostasis remain largely unknown. We used basic concepts of control theory to build a framework for classifying possible components of the homeostatic machinery that stabilizes firing rate, and we particularly emphasize the potential role of sleep and wakefulness in this homeostatic process. This framework may facilitate the identification of new homeostatic pathways whose malfunctions drive instability of neural circuits in distinct brain disorders.
Collapse
Affiliation(s)
- Antonella Ruggiero
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Maxim Katsenelson
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel.
| |
Collapse
|
20
|
Huo Q, Tabassum S, Chen M, Sun M, Deng Y, Zheng X, Li Y, Chen J, Long C, Yang L. Amyloid-β Protein Precursor Deficiency Changes Neuronal Electrical Activity and Levels of Mitochondrial Proteins in the Medial Prefrontal Cortex. J Alzheimers Dis 2021; 81:1469-1482. [PMID: 33935084 DOI: 10.3233/jad-201557] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Neuropathological features of Alzheimer's disease are characterized by the deposition of amyloid-β (Aβ) plaques and impairments in synaptic activity and memory. However, we know little about the physiological role of amyloid-β protein precursor (AβPP) from which Aβ derives. OBJECTIVE Evaluate APP deficiency induced alterations in neuronal electrical activity and mitochondrial protein expression. METHODS Utilizing electrophysiological, biochemical, pharmacological, and behavioral tests, we revealed aberrant local field potential (LFP), extracellular neuronal firing and levels of mitochondrial proteins. RESULT We show that APP knockout (APP-/-) leads to increased gamma oscillations in the medial prefrontal cortex (mPFC) at 1-2 months old, which can be restored by baclofen (Bac), a γ-aminobutyric acid type B receptor (GABABR) agonist. A higher dose and longer exposure time is required for Bac to suppress neuronal firing in APP-/- mice than in wild type animals, indicating enhanced GABABR mediated activity in the mPFC of APP-/- mice. In line with increased GABABR function, the glutamine synthetase inhibitor, L-methionine sulfonate, significantly increases GABABR levels in the mPFC of APP-/- mice and this is associated with a significantly lower incidence of death. The results suggest that APP-/- mice developed stronger GABABR mediated inhibition. Using HEK 293 as an expression system, we uncover that AβPP functions to suppress GABABR expression. Furthermore, APP-/- mice show abnormal expression of several mitochondrial proteins. CONCLUSION APP deficiency leads to both abnormal network activity involving defected GABABR and mitochondrial dysfunction, suggesting critical role of AβPP in synaptic and network function.
Collapse
Affiliation(s)
- Qingwei Huo
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, China
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sidra Tabassum
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Ming Chen
- Department of Pharmacology, Key Laboratory of Anti-inflammatory and Immunopharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Mengyao Sun
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Yueming Deng
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Xingzhi Zheng
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Yi Li
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Jian Chen
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Cheng Long
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Li Yang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, China
| |
Collapse
|
21
|
Salazar AM, Leisgang AM, Ortiz AA, Murtishaw AS, Kinney JW. Alterations of GABA B receptors in the APP/PS1 mouse model of Alzheimer's disease. Neurobiol Aging 2020; 97:129-143. [PMID: 33232936 DOI: 10.1016/j.neurobiolaging.2020.10.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 09/08/2020] [Accepted: 10/16/2020] [Indexed: 01/20/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the progressive decline of memory and cognitive function. The disease is characterized by the presence of amyloid plaques, tau tangles, altered inflammatory signaling, and alterations in numerous neurotransmitter signaling systems, including γ-aminobutyric acid (GABA). Given the extensive role of GABA in regulating neuronal activity, a careful investigation of GABA-related changes is needed. Further, given persistent inflammation has been demonstrated to drive AD pathology, the presence of GABA B receptor expressed on glia that serve a role regulation of the immune response adds to potential implications of altered GABA in AD. There has not previously been a systematic evaluation of GABA-related changes in an amyloid model of AD that specifically focuses on examining changes in GABA B receptors. In the present study, we examined alterations in several GABA-specific targets in the APP/PS1 mouse model at different ages. In the 4-month-old cohort, no significant deficits in spatial learning and memory or alterations in any of the GABAergic targets were observed compared with wild-type controls. However, we identified significant alterations in several GABA-related targets in the 6-month-old cohort that exhibited spatial learning deficits that include changes in glutamic acid decarboxylase 65, GABA transporter type 3, and GABA B receptors protein and mRNA levels. This was the same cohort at which learning and memory deficits and significant amyloid pathology was observed. Overall, our study provides evidence of altered GABAergic signaling in an amyloid model of AD at a time point consistent with AD-related deficits.
Collapse
Affiliation(s)
- Arnold M Salazar
- Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Amanda M Leisgang
- Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Andrew A Ortiz
- Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Andrew S Murtishaw
- Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Jefferson W Kinney
- Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA.
| |
Collapse
|
22
|
Mechanisms and Regulation of Neuronal GABA B Receptor-Dependent Signaling. Curr Top Behav Neurosci 2020; 52:39-79. [PMID: 32808092 DOI: 10.1007/7854_2020_129] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
γ-Aminobutyric acid B receptors (GABABRs) are broadly expressed throughout the central nervous system where they play an important role in regulating neuronal excitability and synaptic transmission. GABABRs are G protein-coupled receptors that mediate slow and sustained inhibitory actions via modulation of several downstream effector enzymes and ion channels. GABABRs are obligate heterodimers that associate with diverse arrays of proteins to form modular complexes that carry out distinct physiological functions. GABABR-dependent signaling is fine-tuned and regulated through a multitude of mechanisms that are relevant to physiological and pathophysiological states. This review summarizes the current knowledge on GABABR signal transduction and discusses key factors that influence the strength and sensitivity of GABABR-dependent signaling in neurons.
Collapse
|
23
|
Abstract
Baclofen, β-(4-chlorophenyl)-γ-aminobutyric acid, holds a unique position in neuroscience, remaining the only U.S. Food and Drug Administration (FDA) approved GABAB agonist. While intended to be a more brain penetrant, i.e, ability to cross the blood-brain barrier (BBB), version of GABA (γ-aminobutyric acid) for the potential treatment of epilepsy, baclofen's highly efficacious muscle relaxant properties led to its approval, as a racemate, for the treatment of spasticity. Interestingly, baclofen received FDA approval before its receptor, GABAB, was discovered and its exact mechanism of action was known. In recent times, baclofen has a myriad of off-label uses, with the treatment for alcohol abuse and drug addiction garnering a great deal of attention. This Review aims to capture the >60 year legacy of baclofen by walking through the history, pharmacology, synthesis, drug metabolism, routes of administration, and societal impact of this Classic in chemical neuroscience.
Collapse
Affiliation(s)
- Caitlin N. Kent
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Chemistry, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Charlotte Park
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Craig W. Lindsley
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Chemistry, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
24
|
M-Current Inhibition in Hippocampal Excitatory Neurons Triggers Intrinsic and Synaptic Homeostatic Responses at Different Temporal Scales. J Neurosci 2020; 40:3694-3706. [PMID: 32277041 DOI: 10.1523/jneurosci.1914-19.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 11/21/2022] Open
Abstract
Persistent alterations in neuronal activity elicit homeostatic plastic changes in synaptic transmission and/or intrinsic excitability. However, it is unknown whether these homeostatic processes operate in concert or at different temporal scales to maintain network activity around a set-point value. Here we show that chronic neuronal hyperactivity, induced by M-channel inhibition, triggered intrinsic and synaptic homeostatic plasticity at different timescales in cultured hippocampal pyramidal neurons from mice of either sex. Homeostatic changes of intrinsic excitability occurred at a fast timescale (1-4 h) and depended on ongoing spiking activity. This fast intrinsic adaptation included plastic changes in the threshold current and a distal relocation of FGF14, a protein physically bridging Nav1.6 and Kv7.2 channels along the axon initial segment. In contrast, synaptic adaptations occurred at a slower timescale (∼2 d) and involved decreases in miniature EPSC amplitude. To examine how these temporally distinct homeostatic responses influenced hippocampal network activity, we quantified the rate of spontaneous spiking measured by multielectrode arrays at extended timescales. M-Channel blockade triggered slow homeostatic renormalization of the mean firing rate (MFR), concomitantly accompanied by a slow synaptic adaptation. Thus, the fast intrinsic adaptation of excitatory neurons is not sufficient to account for the homeostatic normalization of the MFR. In striking contrast, homeostatic adaptations of intrinsic excitability and spontaneous MFR failed in hippocampal GABAergic inhibitory neurons, which remained hyperexcitable following chronic M-channel blockage. Our results indicate that a single perturbation such as M-channel inhibition triggers multiple homeostatic mechanisms that operate at different timescales to maintain network mean firing rate.SIGNIFICANCE STATEMENT Persistent alterations in synaptic input elicit homeostatic plastic changes in neuronal activity. Here we show that chronic neuronal hyperexcitability, induced by M-type potassium channel inhibition, triggered intrinsic and synaptic homeostatic plasticity at different timescales in hippocampal excitatory neurons. The data indicate that the fast adaptation of intrinsic excitability depends on ongoing spiking activity but is not sufficient to provide homeostasis of the mean firing rate. Our results show that a single perturbation such as M-channel inhibition can trigger multiple homeostatic processes that operate at different timescales to maintain network mean firing rate.
Collapse
|
25
|
Negri J, Menon V, Young-Pearse TL. Assessment of Spontaneous Neuronal Activity In Vitro Using Multi-Well Multi-Electrode Arrays: Implications for Assay Development. eNeuro 2020; 7:ENEURO.0080-19.2019. [PMID: 31896559 PMCID: PMC6984810 DOI: 10.1523/eneuro.0080-19.2019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022] Open
Abstract
Multi-electrode arrays (MEAs) are being more widely used by researchers as an instrument platform for monitoring prolonged, non-destructive recordings of spontaneously firing neurons in vitro for applications in modeling Alzheimer's, Parkinson's, schizophrenia, and many other diseases of the human CNS. With the more widespread use of these instruments, there is a need to examine the prior art of studies utilizing MEAs and delineate best practices for data acquisition and analysis to avoid errors in interpretation of the resultant data. Using a dataset of recordings from primary rat (Rattus norvegicus) cortical cultures, methods and statistical power for discerning changes in neuronal activity on the array level are examined. Further, a method for unsupervised spike sorting is implemented, allowing for the resolution of action potential incidents down to the single neuron level. Following implementation of spike sorting, the dynamics of firing frequency across populations of individual neurons and networks are examined longitudinally. Finally, the ability to detect a frequency independent phenotype, the change in action potential amplitude, is demonstrated through the use of pore-forming neurotoxin treatments. Taken together, this study provides guidance and tools for users wishing to incorporate multi-well MEA usage into their studies.
Collapse
Affiliation(s)
- Joseph Negri
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
- Graduate Program in Biological and Biomedical Sciences, Division of Medical Sciences, Harvard University, Cambridge, MA 02138
| | - Vilas Menon
- Department of Neurology, Columbia University Medical Center, New York, NY 10032
| | - Tracy L Young-Pearse
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
26
|
Frere S, Slutsky I. Alzheimer's Disease: From Firing Instability to Homeostasis Network Collapse. Neuron 2019; 97:32-58. [PMID: 29301104 DOI: 10.1016/j.neuron.2017.11.028] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/14/2017] [Accepted: 11/17/2017] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) starts from pure cognitive impairments and gradually progresses into degeneration of specific brain circuits. Although numerous factors initiating AD have been extensively studied, the common principles underlying the transition from cognitive deficits to neuronal loss remain unknown. Here we describe an evolutionarily conserved, integrated homeostatic network (IHN) that enables functional stability of central neural circuits and safeguards from neurodegeneration. We identify the critical modules comprising the IHN and propose a central role of neural firing in controlling the complex homeostatic network at different spatial scales. We hypothesize that firing instability and impaired synaptic plasticity at early AD stages trigger a vicious cycle, leading to dysregulation of the whole IHN. According to this hypothesis, the IHN collapse represents the major driving force of the transition from early memory impairments to neurodegeneration. Understanding the core elements of homeostatic control machinery, the reciprocal connections between distinct IHN modules, and the role of firing homeostasis in this hierarchy has important implications for physiology and should offer novel conceptual approaches for AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Samuel Frere
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel.
| |
Collapse
|
27
|
Tang BL. Amyloid Precursor Protein (APP) and GABAergic Neurotransmission. Cells 2019; 8:E550. [PMID: 31174368 PMCID: PMC6627941 DOI: 10.3390/cells8060550] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/25/2019] [Accepted: 06/06/2019] [Indexed: 12/16/2022] Open
Abstract
The amyloid precursor protein (APP) is the parent polypeptide from which amyloid-beta (Aβ) peptides, key etiological agents of Alzheimer's disease (AD), are generated by sequential proteolytic processing involving β- and γ-secretases. APP mutations underlie familial, early-onset AD, and the involvement of APP in AD pathology has been extensively studied. However, APP has important physiological roles in the mammalian brain, particularly its modulation of synaptic functions and neuronal survival. Recent works have now shown that APP could directly modulate γ-aminobutyric acid (GABA) neurotransmission in two broad ways. Firstly, APP is shown to interact with and modulate the levels and activity of the neuron-specific Potassium-Chloride (K+-Cl-) cotransporter KCC2/SLC12A5. The latter is key to the maintenance of neuronal chloride (Cl-) levels and the GABA reversal potential (EGABA), and is therefore important for postsynaptic GABAergic inhibition through the ionotropic GABAA receptors. Secondly, APP binds to the sushi domain of metabotropic GABAB receptor 1a (GABABR1a). In this regard, APP complexes and is co-transported with GABAB receptor dimers bearing GABABR1a to the axonal presynaptic plasma membrane. On the other hand, secreted (s)APP generated by secretase cleavages could act as a GABABR1a-binding ligand that modulates presynaptic vesicle release. The discovery of these novel roles and activities of APP in GABAergic neurotransmission underlies the physiological importance of APP in postnatal brain function.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117597, Singapore.
| |
Collapse
|
28
|
Styr B, Gonen N, Zarhin D, Ruggiero A, Atsmon R, Gazit N, Braun G, Frere S, Vertkin I, Shapira I, Harel M, Heim LR, Katsenelson M, Rechnitz O, Fadila S, Derdikman D, Rubinstein M, Geiger T, Ruppin E, Slutsky I. Mitochondrial Regulation of the Hippocampal Firing Rate Set Point and Seizure Susceptibility. Neuron 2019; 102:1009-1024.e8. [PMID: 31047779 PMCID: PMC6559804 DOI: 10.1016/j.neuron.2019.03.045] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 02/07/2019] [Accepted: 03/28/2019] [Indexed: 01/08/2023]
Abstract
Maintaining average activity within a set-point range constitutes a fundamental property of central neural circuits. However, whether and how activity set points are regulated remains unknown. Integrating genome-scale metabolic modeling and experimental study of neuronal homeostasis, we identified mitochondrial dihydroorotate dehydrogenase (DHODH) as a regulator of activity set points in hippocampal networks. The DHODH inhibitor teriflunomide stably suppressed mean firing rates via synaptic and intrinsic excitability mechanisms by modulating mitochondrial Ca2+ buffering and spare respiratory capacity. Bi-directional activity perturbations under DHODH blockade triggered firing rate compensation, while stabilizing firing to the lower level, indicating a change in the firing rate set point. In vivo, teriflunomide decreased CA3-CA1 synaptic transmission and CA1 mean firing rate and attenuated susceptibility to seizures, even in the intractable Dravet syndrome epilepsy model. Our results uncover mitochondria as a key regulator of activity set points, demonstrate the differential regulation of set points and compensatory mechanisms, and propose a new strategy to treat epilepsy.
Collapse
Affiliation(s)
- Boaz Styr
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Nir Gonen
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Daniel Zarhin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Antonella Ruggiero
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Refaela Atsmon
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Neta Gazit
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Gabriella Braun
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Samuel Frere
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Irena Vertkin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Ilana Shapira
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Michal Harel
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Leore R Heim
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Maxim Katsenelson
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Ohad Rechnitz
- Department of Neuroscience, Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, 31096 Haifa, Israel
| | - Saja Fadila
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; The Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Dori Derdikman
- Department of Neuroscience, Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, 31096 Haifa, Israel
| | - Moran Rubinstein
- Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel; Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; The Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Tamar Geiger
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Eytan Ruppin
- Cancer Data Science Lab (CDSL), National Cancer Institute, NIH, Bethesda, MD, USA
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel.
| |
Collapse
|
29
|
Dubovyk V, Manahan-Vaughan D. Distinct Time-Course of Alterations of Groups I and II Metabotropic Glutamate Receptor and GABAergic Receptor Expression Along the Dorsoventral Hippocampal Axis in an Animal Model of Psychosis. Front Behav Neurosci 2019; 13:98. [PMID: 31139061 PMCID: PMC6519509 DOI: 10.3389/fnbeh.2019.00098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/23/2019] [Indexed: 01/13/2023] Open
Abstract
Psychosis is a clinical state that encompasses a range of abnormal conditions, including distortions in sensory information processing and the resultant delusional thinking, emotional discordance and cognitive impairments. Upon developing this condition, the rate at which cognitive and behavioral deteriorations progress steadily increases suggesting an active contribution of the first psychotic event to the progression of structural and functional abnormalities and disease establishment in diagnosed patients. Changes in GABAergic and glutamatergic function, or expression, in the hippocampus have been proposed as a key factor in the pathophysiology of psychosis. However, little is known as to the time-point of onset of putative changes, to what extent they are progressive, and their relation to disease stabilization. Here, we characterized the expression and distribution patterns of groups I and II metabotropic glutamate (mGlu) receptors and GABA receptors 1 week and 3 months after systemic treatment with an N-methyl-D-aspartate receptor (NMDAR) antagonist (MK801) that is used to model a psychosis-like state in adult rats. We found an early alteration in the expression of mGlu1, mGlu2/3, and GABAB receptors across the hippocampal dorsoventral and transverse axes. This expanded to include an up-regulation of mGlu5 levels across the entire CA1 region and a reduction in GABAB expression, as well as GAD67-positive interneurons particularly in the dorsal hippocampus that appeared 3 months after treatment. Our findings indicate that a reduction of excitability may occur in the hippocampus soon after first-episode psychosis. This changes, over time, into increased excitability. These hippocampus-specific alterations are likely to contribute to the pathophysiology and stabilization of psychosis.
Collapse
Affiliation(s)
- Valentyna Dubovyk
- Department of Neurophysiology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany.,International Graduate School of Neuroscience, Ruhr-University Bochum, Bochum, Germany
| | | |
Collapse
|
30
|
Gottshall JL, Adams ZM, Forgacs PB, Schiff ND. Daytime Central Thalamic Deep Brain Stimulation Modulates Sleep Dynamics in the Severely Injured Brain: Mechanistic Insights and a Novel Framework for Alpha-Delta Sleep Generation. Front Neurol 2019; 10:20. [PMID: 30778326 PMCID: PMC6369150 DOI: 10.3389/fneur.2019.00020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/08/2019] [Indexed: 12/17/2022] Open
Abstract
Loss of organized sleep electrophysiology is a characteristic finding following severe brain injury. The return of structured elements of sleep architecture has been associated with positive prognosis across injury etiologies, suggesting a role for sleep dynamics as biomarkers of wakeful neuronal circuit function. In a continuing study of one minimally conscious state patient studied over the course of ~8½ years, we sought to investigate whether changes in daytime brain activation induced by central thalamic deep brain stimulation (CT-DBS) influenced sleep electrophysiology. In this patient subject, we previously reported significant improvements in sleep electrophysiology during 5½ years of CT-DBS treatment, including increased sleep spindle frequency and SWS delta power. We now present novel findings that many of these improvements in sleep electrophysiology regress following CT-DBS discontinuation; these regressions in sleep features correlate with a significant decrease in behavioral responsiveness. We also observe the re-emergence of alpha-delta sleep, which had been previously suppressed by daytime CT-DBS in this patient subject. Importantly, CT-DBS was only active during the daytime and has been proposed to mediate recovery of consciousness by driving synaptic activity across frontostriatal systems through the enhancement of thalamocortical output. Accordingly, the improvement of sleep dynamics during daytime CT-DBS and their subsequent regression following CT-DBS discontinuation implicates wakeful synaptic activity as a robust modulator of sleep electrophysiology. We interpret these findings in the context of the “synaptic homeostasis hypothesis,” whereby we propose that daytime upregulation of thalamocortical output in the severely injured brain may facilitate organized frontocortical circuit activation and yield net synaptic potentiation during wakefulness, providing a homeostatic drive that reconstitutes sleep dynamics over time. Furthermore, we consider common large-scale network dynamics across several neuropsychiatric disorders in which alpha-delta sleep has been documented, allowing us to formulate a novel mechanistic framework for alpha-delta sleep generation. We conclude that the bi-directional modulation of sleep electrophysiology by daytime thalamocortical activity in the severely injured brain: (1) emphasizes the cyclical carry-over effects of state-dependent circuit activation on large-scale brain dynamics, and (2) further implicates sleep electrophysiology as a sensitive indicator of wakeful brain activation and covert functional recovery in the severely injured brain.
Collapse
Affiliation(s)
- Jackie L Gottshall
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Zoe M Adams
- Department of Neurology, Weill Cornell Medicine, New York, NY, United States
| | - Peter B Forgacs
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States.,Department of Neurology, Weill Cornell Medicine, New York, NY, United States.,Rockefeller University Hospital, New York, NY, United States
| | - Nicholas D Schiff
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States.,Department of Neurology, Weill Cornell Medicine, New York, NY, United States.,Rockefeller University Hospital, New York, NY, United States
| |
Collapse
|
31
|
Rice HC, de Malmazet D, Schreurs A, Frere S, Van Molle I, Volkov AN, Creemers E, Vertkin I, Nys J, Ranaivoson FM, Comoletti D, Savas JN, Remaut H, Balschun D, Wierda KD, Slutsky I, Farrow K, De Strooper B, de Wit J. Secreted amyloid-β precursor protein functions as a GABA BR1a ligand to modulate synaptic transmission. Science 2019; 363:eaao4827. [PMID: 30630900 PMCID: PMC6366617 DOI: 10.1126/science.aao4827] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/30/2018] [Accepted: 11/14/2018] [Indexed: 12/16/2022]
Abstract
Amyloid-β precursor protein (APP) is central to the pathogenesis of Alzheimer's disease, yet its physiological function remains unresolved. Accumulating evidence suggests that APP has a synaptic function mediated by an unidentified receptor for secreted APP (sAPP). Here we show that the sAPP extension domain directly bound the sushi 1 domain specific to the γ-aminobutyric acid type B receptor subunit 1a (GABABR1a). sAPP-GABABR1a binding suppressed synaptic transmission and enhanced short-term facilitation in mouse hippocampal synapses via inhibition of synaptic vesicle release. A 17-amino acid peptide corresponding to the GABABR1a binding region within APP suppressed in vivo spontaneous neuronal activity in the hippocampus of anesthetized Thy1-GCaMP6s mice. Our findings identify GABABR1a as a synaptic receptor for sAPP and reveal a physiological role for sAPP in regulating GABABR1a function to modulate synaptic transmission.
Collapse
Affiliation(s)
- Heather C Rice
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Daniel de Malmazet
- Neuro-Electronics Research Flanders, Leuven, Belgium
- Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - An Schreurs
- Brain & Cognition, KU Leuven, Leuven, Belgium
| | - Samuel Frere
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Inge Van Molle
- VIB-VUB Structural Biology Research Center, Brussels, Belgium
| | - Alexander N Volkov
- VIB-VUB Structural Biology Research Center, Brussels, Belgium
- Jean Jeener NMR Centre, VUB, Brussels, Belgium
| | - Eline Creemers
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Irena Vertkin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Julie Nys
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Fanomezana M Ranaivoson
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, NJ, USA
| | - Davide Comoletti
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, NJ, USA
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, NJ, USA
| | - Jeffrey N Savas
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Han Remaut
- VIB-VUB Structural Biology Research Center, Brussels, Belgium
| | | | - Keimpe D Wierda
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Karl Farrow
- Neuro-Electronics Research Flanders, Leuven, Belgium
- Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- VIB, Leuven, Belgium
- imec, Leuven, Belgium
| | - Bart De Strooper
- VIB Center for Brain & Disease Research, Leuven, Belgium.
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- UK-Dementia Research Institute at University College London, UK
| | - Joris de Wit
- VIB Center for Brain & Disease Research, Leuven, Belgium.
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
32
|
Li Y, Liu Y, Fan J, Zhou Q, Song X, Peng Z, Qin Z, Tao T. Validation and bioinformatic analysis of propofol-induced differentially expressed microRNAs in primary cultured neural stem cells. Gene 2018; 664:90-100. [PMID: 29679758 DOI: 10.1016/j.gene.2018.04.046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/26/2018] [Accepted: 04/16/2018] [Indexed: 11/27/2022]
Abstract
Propofol, a widely used intravenous anesthetic, was previously considered as a neuroprotective agent. Recently, however, accumulating evidence suggests that it may cause neurotoxicity, especially in the development of neural stem cells (NSCs). The potential mechanisms contributing to propofol-induced neurotoxicity during neurogenesis, such as those involving microRNAs (miRNAs), are still unknown. In this study, a total of 27 differentially expressed miRNAs were identified in our initial screen and 6 miRNAs were validated by qRT-PCR. Three miRNAs were up-regulated (miR-377-5p, miR-194-3p and miR-143-5p), and three were down-regulated (miR-3583-3p, miR-466b-5p and miR-410-5p). Following gene ontology and KEGG pathway enrichment analysis, Gabbr1, Canca1b and Gabbr2, which are enriched in the GABAergic synapse pathway, were selected as genes potentially playing a role in propofol-induced neurotoxicity. Gabbr1 and Cacna1b, which are targeted by miRNAs that are up-regulated following propofol exposure, showed decreased expression at the mRNA and protein levels. Gabbr2, targeted by miRNAs that were down-regulated following treatment with propofol, was up-regulated at both the levels of mRNA and protein expression. The two clusters of miRNAs that show differential expression following propofol exposure may act in a synergistic manner to regulate several genes simultaneously during the development of NSCs. Our results may contribute to clarify the molecular mechanism and provide potential therapeutic targets for propofol induced neurotoxicity.
Collapse
Affiliation(s)
- Yan Li
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Youtan Liu
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Jun Fan
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Key Laboratory of Psychiatric Disorders of Guangdong Province, Guangzhou, China
| | - Quan Zhou
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiuling Song
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiyong Peng
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Zaisheng Qin
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Tao Tao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
33
|
Jacobson LH, Vlachou S, Slattery DA, Li X, Cryan JF. The Gamma-Aminobutyric Acid B Receptor in Depression and Reward. Biol Psychiatry 2018; 83:963-976. [PMID: 29759132 DOI: 10.1016/j.biopsych.2018.02.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 01/14/2018] [Accepted: 02/06/2018] [Indexed: 12/31/2022]
Abstract
The metabotropic gamma-aminobutyric acid B (GABAB) receptor was the first described obligate G protein-coupled receptor heterodimer and continues to set the stage for discoveries in G protein-coupled receptor signaling complexity. In this review, dedicated to the life and work of Athina Markou, we explore the role of GABAB receptors in depression, reward, and the convergence of these domains in anhedonia, a shared symptom of major depressive disorder and withdrawal from drugs of abuse. GABAB receptor expression and function are enhanced by antidepressants and reduced in animal models of depression. Generally, GABAB receptor antagonists are antidepressant-like and agonists are pro-depressive. Exceptions to this rule likely reflect the differential influence of GABAB1 isoforms in depression-related behavior and neurobiology, including the anhedonic effects of social stress. A wealth of data implicate GABAB receptors in the rewarding effects of drugs of abuse. We focus on nicotine as an example. GABAB receptor activation attenuates, and deactivation enhances, nicotine reward and associated neurobiological changes. In nicotine withdrawal, however, GABAB receptor agonists, antagonists, and positive allosteric modulators enhance anhedonia, perhaps owing to differential effects of GABAB1 isoforms on the dopaminergic system. Nicotine cue-induced reinstatement is more reliably attenuated by GABAB receptor activation. Separation of desirable and undesirable side effects of agonists is achievable with positive allosteric modulators, which are poised to enter clinical studies for drug abuse. GABAB1 isoforms are key to understanding the neurobiology of anhedonia, whereas allosteric modulators may offer a mechanism for targeting specific brain regions and processes associated with reward and depression.
Collapse
Affiliation(s)
- Laura H Jacobson
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia; Department of Pharmacology and Therapeutics, University of Melbourne, Victoria, Australia.
| | - Styliani Vlachou
- School of Nursing and Human Sciences, Faculty of Science and Health, Dublin City University, Glasnevin, Dublin
| | - David A Slattery
- Klinik für Psychiatrie, Psychosomatik und Psychotherapie, Universitätsklinikum Frankfurt, Frankfurt am Main, Germany
| | - Xia Li
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, California
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| |
Collapse
|
34
|
Moser A, Hanssen H, Wandinger KP. Excessively increased CSF glutamate levels in GABA B-receptor antibody associated encephalitis: A case report. J Neurol Sci 2018; 388:10-11. [PMID: 29627000 DOI: 10.1016/j.jns.2018.02.041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 02/20/2018] [Accepted: 02/25/2018] [Indexed: 11/30/2022]
Affiliation(s)
- Andreas Moser
- Department of Neurology, University of Luebeck, Luebeck, Germany; Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, Freiburg, Germany
| | - Henrike Hanssen
- Department of Neurology, University of Luebeck, Luebeck, Germany; Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - Klaus-Peter Wandinger
- Department of Neurology, University of Luebeck, Luebeck, Germany; Institute of Clinical Chemistry, University Hospital of Schleswig-Holstein, Luebeck, Germany.
| |
Collapse
|
35
|
Styr B, Slutsky I. Imbalance between firing homeostasis and synaptic plasticity drives early-phase Alzheimer's disease. Nat Neurosci 2018; 21:463-473. [PMID: 29403035 DOI: 10.1038/s41593-018-0080-x] [Citation(s) in RCA: 219] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 01/08/2018] [Indexed: 12/18/2022]
Abstract
During recent years, the preclinical stage of Alzheimer's disease (AD) has become a major focus of research. Continued failures in clinical trials and the realization that early intervention may offer better therapeutic outcome triggered a conceptual shift from late-stage AD pathology to early-stage pathophysiology. While much effort has been directed at understanding the factors initiating AD, little is known about the principle basis underlying the disease progression at its early stages. In this Perspective, we suggest a hypothesis to explain the transition from 'silent' signatures of aberrant neural circuit activity to clinically evident memory impairments. Namely, we propose that failures in firing homeostasis and imbalance between firing stability and synaptic plasticity in cortico-hippocampal circuits represent the driving force of early disease progression. We analyze the main types of possible homeostatic failures and provide the essential conceptual framework for examining the causal link between dysregulation of firing homeostasis, aberrant neural circuit activity and memory-related plasticity impairments associated with early AD.
Collapse
Affiliation(s)
- Boaz Styr
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel. .,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
36
|
The Dual Function of the Polybasic Juxtamembrane Region of Syntaxin 1A in Clamping Spontaneous Release and Stimulating Ca 2+-Triggered Release in Neuroendocrine Cells. J Neurosci 2017; 38:220-231. [PMID: 29133430 DOI: 10.1523/jneurosci.1541-17.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 09/07/2017] [Accepted: 10/07/2017] [Indexed: 11/21/2022] Open
Abstract
The exact function of the polybasic juxtamembrane region (5RK) of the plasma membrane neuronal SNARE, syntaxin 1A (Syx), in vesicle exocytosis, although widely studied, is currently not clear. Here, we addressed the role of 5RK in Ca2+-triggered release, using our Syx-based intramolecular fluorescence resonance energy transfer (FRET) probe, which previously allowed us to resolve a depolarization-induced Ca2+-dependent close-to-open transition (CDO) of Syx that occurs concomitant with evoked release, both in PC12 cells and hippocampal neurons and was abolished upon charge neutralization of 5RK. First, using dynamic FRET analysis in PC12 cells, we show that CDO occurs following assembly of SNARE complexes that include the vesicular SNARE, synaptobrevin 2, and that the participation of 5RK in CDO goes beyond its participation in the final zippering of the complex, because mutations of residues adjacent to 5RK, believed to be crucial for final zippering, do not abolish this transition. In addition, we show that CDO is contingent on membrane phosphatidylinositol 4,5-bisphosphate (PIP2), which is fundamental for maintaining regulated exocytosis, as depletion of membranal PIP2 abolishes CDO. Prompted by these results, which underscore a potentially significant role of 5RK in exocytosis, we next amperometrically analyzed catecholamine release from PC12 cells, revealing that charge neutralization of 5RK promotes spontaneous and inhibits Ca2+-triggered release events. Namely, 5RK acts as a fusion clamp, making release dependent on stimulation by Ca2+SIGNIFICANCE STATEMENT Syntaxin 1A (Syx) is a central protein component of the SNARE complex, which underlies neurotransmitter release. Although widely studied in relation to its participation in SNARE complex formation and its interaction with phosphoinositides, the function of Syx's polybasic juxtamembrane region (5RK) remains unclear. Previously, we showed that a conformational transition of Syx, related to calcium-triggered release, reported by a Syx-based FRET probe, is abolished upon charge neutralization of 5RK (5RK/A). Here we show that this conformational transition is dependent on phosphatidylinositol 4,5-bisphosphate (PIP2) and is related to SNARE complex formation. Subsequently, we show that the 5RK/A mutation enhances spontaneous release and inhibits calcium-triggered release in neuroendocrine cells, indicating a previously unrecognized role of 5RK in neurotransmitter release.
Collapse
|
37
|
Human Brain-Derived Aβ Oligomers Bind to Synapses and Disrupt Synaptic Activity in a Manner That Requires APP. J Neurosci 2017; 37:11947-11966. [PMID: 29101243 DOI: 10.1523/jneurosci.2009-17.2017] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 09/19/2017] [Accepted: 09/29/2017] [Indexed: 12/20/2022] Open
Abstract
Compelling genetic evidence links the amyloid precursor protein (APP) to Alzheimer's disease (AD) and several theories have been advanced to explain the relationship. A leading hypothesis proposes that a small amphipathic fragment of APP, the amyloid β-protein (Aβ), self-associates to form soluble aggregates that impair synaptic and network activity. Here, we used the most disease-relevant form of Aβ, protein isolated from AD brain. Using this material, we show that the synaptotoxic effects of Aβ depend on expression of APP and that the Aβ-mediated impairment of synaptic plasticity is accompanied by presynaptic effects that disrupt the excitatory/inhibitory (E/I) balance. The net increase in the E/I ratio and inhibition of plasticity are associated with Aβ localizing to synapses and binding of soluble Aβ aggregates to synapses requires the expression of APP. Our findings indicate a role for APP in AD pathogenesis beyond the generation of Aβ and suggest modulation of APP expression as a therapy for AD.SIGNIFICANCE STATEMENT Here, we report on the plasticity-disrupting effects of amyloid β-protein (Aβ) isolated from Alzheimer's disease (AD) brain and the requirement of amyloid precursor protein (APP) for these effects. We show that Aβ-containing AD brain extracts block hippocampal LTP, augment glutamate release probability, and disrupt the excitatory/inhibitory balance. These effects are associated with Aβ localizing to synapses and genetic ablation of APP prevents both Aβ binding and Aβ-mediated synaptic dysfunctions. Our results emphasize the importance of APP in AD and should stimulate new studies to elucidate APP-related targets suitable for pharmacological manipulation.
Collapse
|
38
|
Griskova-Bulanova I, Dapsys K, Melynyte S, Voicikas A, Maciulis V, Andruskevicius S, Korostenskaja M. 40Hz auditory steady-state response in schizophrenia: Sensitivity to stimulation type (clicks versus flutter amplitude-modulated tones). Neurosci Lett 2017; 662:152-157. [PMID: 29051085 DOI: 10.1016/j.neulet.2017.10.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 10/12/2017] [Accepted: 10/16/2017] [Indexed: 01/22/2023]
Abstract
Auditory steady-state response (ASSR) at 40Hz has been proposed as a potential biomarker for schizophrenia. The ASSR studies in patients have used click stimulation or amplitude-modulated tones. However, the sensitivity of 40Hz ASSRs to different stimulation types in the same group of patients has not been previously evaluated. Two stimulation types for ASSRs were tested in this study: (1) 40Hz clicks and (2) flutter-amplitude modulated tones. The mean phase-locking index, evoked amplitude and event-related spectral perturbation values were compared between schizophrenia patients (n=26) and healthy controls (n=20). Both stimulation types resulted in the observation of impaired phase-locking and power measures of late (200-500ms) 40Hz ASSR in patients compared to healthy controls. The early-latency (0-100ms) 40Hz ASSR part was diminished in the schizophrenia group in response to clicks only. The late-latency 40Hz ASSR parameters obtained through different stimulation types correlated in healthy subjects but not in patients. We conclude that flutter amplitude-modulated tone stimulation, due to its potential to reveal late-latency entrainment deficits, is suitable for use in clinical populations. Careful consideration of experimental stimulation settings can contribute to the interpretation of ASSR deficits and utilization as a potential biomarker.
Collapse
Affiliation(s)
| | - Kastytis Dapsys
- Department of Electrophysiological Treatment and Investigation Methods, Vilnius Republican Psychiatric Hospital, Vilnius, Lithuania
| | - Sigita Melynyte
- Institute of Biosciences, Vilnius University, Vilnius, Lithuania
| | | | - Valentinas Maciulis
- Department of Electrophysiological Treatment and Investigation Methods, Vilnius Republican Psychiatric Hospital, Vilnius, Lithuania
| | - Sergejus Andruskevicius
- Department of Electrophysiological Treatment and Investigation Methods, Vilnius Republican Psychiatric Hospital, Vilnius, Lithuania
| | - Milena Korostenskaja
- Milena's Functional Brain Mapping and Brain Computer Interface Lab, Florida Hospital for Children, Orlando, FL, USA; MEG Lab, Florida Hospital for Children, Orlando, FL, USA; Department of Psychology, College of Arts and Sciences, University of North Florida, Jacksonville, FL, USA
| |
Collapse
|
39
|
Nasehi M, Alaghmandan-Motlagh N, Ebrahimi-Ghiri M, Nami M, Zarrindast MR. The interaction between hippocampal GABA-B and cannabinoid receptors upon spatial change and object novelty discrimination memory function. Psychopharmacology (Berl) 2017; 234:3117-3128. [PMID: 28779310 DOI: 10.1007/s00213-017-4688-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 07/08/2017] [Indexed: 12/13/2022]
Abstract
RATIONALE Previous studies have postulated functional links between GABA and cannabinoid systems in the hippocampus. The aim of the present study was to investigate any possible interaction between these systems in spatial change and object novelty discrimination memory consolidation in the dorsal hippocampus (CA1 region) of NMRI mice. METHODS Assessment of the spatial change and object novelty discrimination memory function was carried out in a non-associative task. The experiment comprised mice exposure to an open field containing five objects followed by the examination of their reactivity to object displacement (spatial change) and object substitution (object novelty) after three sessions of habituation. RESULTS Our results showed that the post-training intraperitoneal administration of the higher dose of ACPA (0.02 mg/kg) impaired both spatial change and novelty discrimination memory functions. Meanwhile, the higher dose of GABA-B receptor agonist, baclofen, impaired the spatial change memory by itself. Moreover, the post-training intra-CA1 microinjection of a subthreshold dose of baclofen increased the ACPA effect on spatial change and novelty discrimination memory at a lower and higher dose, respectively. On the other hand, the lower and higher but not mid-level doses of GABA-B receptor antagonist, phaclofen, could reverse memory deficits induced by ACPA. However, phaclofen at its mid-level dose impaired the novelty discrimination memory and whereas the higher dose impaired the spatial change memory. CONCLUSIONS Based on our findings, GABA-B receptors in the CA1 region appear to modulate the ACPA-induced cannabinoid CB1 signaling upon spatial change and novelty discrimination memory functions.
Collapse
Affiliation(s)
- Mohammad Nasehi
- Cognitive and Neuroscience Research Center (CNRC), Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran.
| | | | | | - Mohammad Nami
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.,Neuroscience Laboratory (Brain, Cognition and Behavior), Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad-Reza Zarrindast
- Department of Pharmacology School of Medicine, Tehran University of Medical Sciences, Tehran, Iran. .,Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran. .,School of Cognitive Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran. .,Medical Genomics Research Center, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran. .,Institute for Cognitive Science Studies (ICSS), Tehran, Iran.
| |
Collapse
|
40
|
Homeostatic regulation through GABA and acetylcholine muscarinic receptors of motor trigeminal neurons following sleep deprivation. Brain Struct Funct 2017; 222:3163-3178. [PMID: 28299422 PMCID: PMC5585289 DOI: 10.1007/s00429-017-1392-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 02/20/2017] [Indexed: 12/21/2022]
Abstract
Muscle tone is regulated across sleep-wake states, being maximal in waking, reduced in slow wave sleep (SWS) and absent in paradoxical or REM sleep (PS or REMS). Such changes in tone have been recorded in the masseter muscles and shown to correspond to changes in activity and polarization of the trigeminal motor 5 (Mo5) neurons. The muscle hypotonia and atonia during sleep depend in part on GABA acting upon both GABAA and GABAB receptors (Rs) and acetylcholine (ACh) acting upon muscarinic 2 (AChM2) Rs. Here, we examined whether Mo5 neurons undergo homeostatic regulation through changes in these inhibitory receptors following prolonged activity with enforced waking. By immunofluorescence, we assessed that the proportion of Mo5 neurons positively stained for GABAARs was significantly higher after sleep deprivation (SD, ~65%) than sleep control (SC, ~32%) and that the luminance of the GABAAR fluorescence was significantly higher after SD than SC and sleep recovery (SR). Although, all Mo5 neurons were positively stained for GABABRs and AChM2Rs (100%) in all groups, the luminance of these receptors was significantly higher following SD as compared to SC and SR. We conclude that the density of GABAA, GABAB and AChM2 receptors increases on Mo5 neurons during SD. The increase in these receptors would be associated with increased inhibition in the presence of GABA and ACh and thus a homeostatic down-scaling in the excitability of the Mo5 neurons after prolonged waking and resulting increased susceptibility to muscle hypotonia or atonia along with sleep.
Collapse
|
41
|
Pin JP, Bettler B. Organization and functions of mGlu and GABAB receptor complexes. Nature 2016; 540:60-68. [DOI: 10.1038/nature20566] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 10/21/2016] [Indexed: 02/08/2023]
|
42
|
GABA Receptors on Orexin and Melanin-Concentrating Hormone Neurons Are Differentially Homeostatically Regulated Following Sleep Deprivation. eNeuro 2016; 3:eN-NWR-0077-16. [PMID: 27294196 PMCID: PMC4899679 DOI: 10.1523/eneuro.0077-16.2016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/25/2016] [Accepted: 05/27/2016] [Indexed: 11/21/2022] Open
Abstract
Though overlapping in distribution through the hypothalamus, orexin (Orx) and melanin-concentrating hormone (MCH) neurons play opposite roles in the regulation of sleep-wake states. Orx neurons discharge during waking, whereas MCH neurons discharge during sleep. In the present study, we examined in mice whether GABAA and GABAB receptors (Rs) are present on Orx and MCH neurons and might undergo differential changes as a function of their different activities following sleep deprivation (SD) and sleep recovery (SR). Applying quantitative stereological image analysis to dual-immunofluorescent stained sections, we determined that the proportion of Orx neurons positively immunostained for GABAARs was significantly higher following SD (∼48%) compared with sleep control (SC; ∼24%) and SR (∼27%), and that the luminance of the GABAARs was significantly greater. In contrast, the average proportion of the MCH neurons immunostained for GABAARs was insignificantly lower following SD (∼43%) compared with SC (∼54%) and SR (56%), and the luminance of the GABAARs was significantly less. Although, GABABRs were observed in all Orx and MCH neurons (100%), the luminance of these receptors was differentially altered following SD. The intensity of GABABRs in the Orx neurons was significantly greater after SD than after SC and SR, whereas that in the MCH neurons was significantly less. The present results indicate that GABA receptors undergo dynamic and differential changes in the wake-active Orx neurons and the sleep-active MCH neurons as a function of and homeostatic adjustment to their preceding activity and sleep-wake state.
Collapse
|
43
|
Konstantoudaki X, Chalkiadaki K, Tivodar S, Karagogeos D, Sidiropoulou K. Impaired synaptic plasticity in the prefrontal cortex of mice with developmentally decreased number of interneurons. Neuroscience 2016; 322:333-45. [DOI: 10.1016/j.neuroscience.2016.02.048] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 02/20/2016] [Accepted: 02/22/2016] [Indexed: 01/14/2023]
|
44
|
Jurado-Parras MT, Delgado-García JM, Sánchez-Campusano R, Gassmann M, Bettler B, Gruart A. Presynaptic GABAB Receptors Regulate Hippocampal Synapses during Associative Learning in Behaving Mice. PLoS One 2016; 11:e0148800. [PMID: 26848590 PMCID: PMC4743998 DOI: 10.1371/journal.pone.0148800] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 01/21/2016] [Indexed: 12/28/2022] Open
Abstract
GABAB receptors are the G-protein-coupled receptors for GABA, the main inhibitory neurotransmitter in the central nervous system. Pharmacological activation of GABAB receptors regulates neurotransmission and neuronal excitability at pre- and postsynaptic sites. Electrophysiological activation of GABAB receptors in brain slices generally requires strong stimulus intensities. This raises the question as to whether behavioral stimuli are strong enough to activate GABAB receptors. Here we show that GABAB1a-/- mice, which constitutively lack presynaptic GABAB receptors at glutamatergic synapses, are impaired in their ability to acquire an operant learning task. In vivo recordings during the operant conditioning reveal a deficit in learning-dependent increases in synaptic strength at CA3-CA1 synapses. Moreover, GABAB1a-/- mice fail to synchronize neuronal activity in the CA1 area during the acquisition process. Our results support that activation of presynaptic hippocampal GABAB receptors is important for acquisition of a learning task and for learning-associated synaptic changes and network dynamics.
Collapse
Affiliation(s)
| | | | | | - Martin Gassmann
- Department of Biomedicine, University of Basel, 4056, Basel, Switzerland
| | - Bernhard Bettler
- Department of Biomedicine, University of Basel, 4056, Basel, Switzerland
- * E-mail: (AG); (BB)
| | - Agnès Gruart
- Division of Neurosciences, Pablo de Olavide University, 41013, Seville, Spain
- * E-mail: (AG); (BB)
| |
Collapse
|
45
|
Role of GABA(B) receptors in learning and memory and neurological disorders. Neurosci Biobehav Rev 2016; 63:1-28. [PMID: 26814961 DOI: 10.1016/j.neubiorev.2016.01.007] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 12/31/2015] [Accepted: 01/21/2016] [Indexed: 01/13/2023]
Abstract
Although it is evident from the literature that altered GABAB receptor function does affect behavior, these results often do not correspond well. These differences could be due to the task protocol, animal strain, ligand concentration, or timing of administration utilized. Because several clinical populations exhibit learning and memory deficits in addition to altered markers of GABA and the GABAB receptor, it is important to determine whether altered GABAB receptor function is capable of contributing to the deficits. The aim of this review is to examine the effect of altered GABAB receptor function on synaptic plasticity as demonstrated by in vitro data, as well as the effects on performance in learning and memory tasks. Finally, data regarding altered GABA and GABAB receptor markers within clinical populations will be reviewed. Together, the data agree that proper functioning of GABAB receptors is crucial for numerous learning and memory tasks and that targeting this system via pharmaceuticals may benefit several clinical populations.
Collapse
|