1
|
Shu J, Jiao Y, Wei W, Yan A. Spermidine Inhibits M1 Microglia Polarization in a Mouse Model of Parkinson's Disease and BV2 Cells via NF-κB/STAT-1 Pathway. Brain Behav 2025; 15:e70410. [PMID: 40059454 PMCID: PMC11891262 DOI: 10.1002/brb3.70410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/16/2024] [Accepted: 12/28/2024] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Excessively activated M1 microglia release proinflammatory factors that can cause neuronal death and contribute to the development of Parkinson's disease (PD). Recent research indicates that spermidine, a naturally occurring polyamine, may have anti-inflammatory properties. Nonetheless, the specific role of spermidine in Parkinson's disease, particularly how it affects microglia-driven neuroinflammation and the balance between M1 and M2 polarization, is still not fully understood. METHODS We examined the effects of spermidine on the polarization of M1/M2 microglia in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD and lipopolysaccharide (LPS)-stimulated BV2 cells. Methods like RT-PCR, western blotting, and immunofluorescence were used to examine how spermidine influences the polarization of microglia. RESULTS In vivo, spermidine pretreatment reduced the activation of M1 microglia and encouraged the transformation of microglia into the M2 phenotype in the substantia nigra (SN) of PD mice. Additionally, spermidine decreased the release of inflammatory factors and lessened the death of dopaminergic neurons in the SN of these mice. In vitro, spermidine indirectly protected neurons from death by affecting microglial polarization. Furthermore, spermidine preconditioning led to decreased phosphorylation of NF-κB, STAT1, and p38 MAPK, while enhancing the phosphorylation of STAT6, both in vivo and in vitro. Additionally, we observed that the supernatant from BV2 cells was cultured with SH-SY5Y neurons. The findings revealed that the supernatant from LPS-activated BV2 cells notably reduced the viability of SH-SY5Y cells, as well as the levels of brain-derived neurotrophic factor (BDNF), TrkB, PI3K, and p-AKT. However, these effects were significantly reversed by pretreatment with spermidine. CONCLUSION Our research found that spermidine reduced M1 microglial polarization, partially through the inhibition of the NF-κB, STAT1, and p38 MAPK pathways, and encouraged M2 microglial polarization by activating the STAT6 pathway. This action helped to mitigate neuroinflammation in both the MPTP mouse model of Parkinson's disease and LPS-stimulated BV2 cells. Additionally, spermidine provided indirect neuroprotection by activating BDNF-TrkB-PI3K/AKT signaling pathways.
Collapse
Affiliation(s)
- Jun Shu
- Department of NeurologyHuadong Hospital, Fudan UniversityShanghaiChina
| | - Yuqiong Jiao
- Department of NeurologyHuadong Hospital, Fudan UniversityShanghaiChina
| | - Wenshi Wei
- Department of NeurologyHuadong Hospital, Fudan UniversityShanghaiChina
| | - Aijuan Yan
- Department of NeurologyHuadong Hospital, Fudan UniversityShanghaiChina
| |
Collapse
|
2
|
Williams D. Why so slow? Models of parkinsonian bradykinesia. Nat Rev Neurosci 2024; 25:573-586. [PMID: 38937655 DOI: 10.1038/s41583-024-00830-0] [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: 05/20/2024] [Indexed: 06/29/2024]
Abstract
Bradykinesia, or slowness of movement, is a defining feature of Parkinson disease (PD) and a major contributor to the negative effects on quality of life associated with this disorder and related conditions. A dominant pathophysiological model of bradykinesia in PD has existed for approximately 30 years and has been the basis for the development of several therapeutic interventions, but accumulating evidence has made this model increasingly untenable. Although more recent models have been proposed, they also appear to be flawed. In this Perspective, I consider the leading prior models of bradykinesia in PD and argue that a more functionally related model is required, one that considers changes that disrupt the fundamental process of accurate information transmission. In doing so, I review emerging evidence of network level functional connectivity changes, information transfer dysfunction and potential motor code transmission error and present a novel model of bradykinesia in PD that incorporates this evidence. I hope that this model may reconcile inconsistencies in its predecessors and encourage further development of therapeutic interventions.
Collapse
Affiliation(s)
- David Williams
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.
- Department of Neurology, Whipps Cross University Hospital, Barts Health NHS Trust, London, UK.
| |
Collapse
|
3
|
Chen JL, Kuo CC. Inhibition of resurgent Na + currents by rufinamide. Neuropharmacology 2024; 247:109835. [PMID: 38228283 DOI: 10.1016/j.neuropharm.2024.109835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/29/2023] [Accepted: 12/31/2023] [Indexed: 01/18/2024]
Abstract
Na+ channels are essential for the genesis of action potentials in most neurons. After opening by membrane depolarization, Na+ channels enter a series of inactivated states (e.g. the fast, intermediate, and slow inactivated states; or If, Ii, and Is). The inactivated Na+ channel may recover via the open state upon membrane repolarization, giving rise to "resurgent" Na+ currents which could be critical for densely repetitive or burst discharges. We incubated CHO-K1 cells transfected with human NaV1.7 cDNA and measured resurgent currents with whole-cell patch recordings. We found Ii is the major inactivated state responsible for the genesis of resurgent currents. Rufinamide, in therapeutic concentrations, could selectively bind to Ii to slow the recovery process and dose-dependently inhibit resurgent currents. The other Na+ channel-inhibiting antiseizure medications (ASM), such as phenytoin and lacosamide (selectively binds to If and Is, separately), fail to show a similar inhibitory effect in clinically relevant concentrations. Resurgent currents are decreased with lengthening of the prepulse, presumably because of redistribution of the channel from Ii to If. Rufinamide could accentuate the decrease to mimic a use-dependent inhibitory effect. The molecular action of slowing of recovery from inactivation by binding to Ii also explains the highly correlative inhibitory effect of rufinamide on both transient and resurgent Na+ currents. The modest but correlative inhibition of both currents may make a novel synergistic effect and thus strong-enough suppression of pathological repetitive and especially burst discharges. Rufinamide may thus have a unique spectrum of therapeutic applications for disorders with excessive neural excitabilities.
Collapse
Affiliation(s)
- Jian-Lin Chen
- Department of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Chung-Chin Kuo
- Department of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
4
|
Zhang Y, Ma L, Zhang X, Yue L, Wang J, Zheng J, Cui S, Liu FY, Wang Z, Wan Y, Yi M. Deep brain stimulation in the lateral habenula reverses local neuronal hyperactivity and ameliorates depression-like behaviors in rats. Neurobiol Dis 2023; 180:106069. [PMID: 36893902 DOI: 10.1016/j.nbd.2023.106069] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/22/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Deep brain stimulation (DBS) is a promising therapy for treatment-resistant depression, while mechanisms underlying its therapeutic effects remain poorly defined. Increasing evidence has revealed an intimate association between the lateral habenula (LHb) and major depression, and suggests that the LHb might be an effective target of DBS therapy for depression. Here, we found that DBS in the LHb effectively decreased depression-like behaviors in rats experienced with chronic unpredictable mild stress (CUMS), a well-accepted paradigm for modeling depression in rodents. In vivo electrophysiological recording unveiled that CUMS increased neuronal burst firing, as well as the proportion of neurons showing hyperactivity to aversive stimuli in the LHb. Nevertheless, DBS downregulated local field potential power, reversed the CUMS-induced increase of LHb burst firing and neuronal hyperactivity to aversive stimuli, and decreased the coherence between LHb and ventral tegmental area (VTA). Our results demonstrate that DBS in the LHb exerts antidepressant-like effects and reverses local neural hyperactivity, supporting the LHb as a target of DBS therapy for depression.
Collapse
Affiliation(s)
- Yuqi Zhang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100083, PR China
| | - Longyu Ma
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100083, PR China
| | - Xueying Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Lupeng Yue
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Science, Beijing 100101, China
| | - Jiaxin Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100083, PR China
| | - Jie Zheng
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100083, PR China
| | - Shuang Cui
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100083, PR China
| | - Feng-Yu Liu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100083, PR China
| | - Zhiyan Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Science, Beijing 100101, China; National Engineering Laboratory for Neuromodulation, School of Aerospace Engineering, Tsinghua University, Beijing 100084, China.
| | - You Wan
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100083, PR China; Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing 100083, PR China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.
| | - Ming Yi
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100083, PR China; Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing 100083, PR China.
| |
Collapse
|
5
|
Sun S, Wang X, Shi X, Fang H, Sun Y, Li M, Han H, He Q, Wang X, Zhang X, Zhu ZW, Chen F, Wang M. Neural pathway connectivity and discharge changes between M1 and STN in hemiparkinsonian rats. Brain Res Bull 2023; 196:1-19. [PMID: 36878325 DOI: 10.1016/j.brainresbull.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/27/2023] [Accepted: 03/03/2023] [Indexed: 03/07/2023]
Abstract
Alterations of electrophysiological activities, such as changed spike firing rates, reshaping the firing patterns, and aberrant frequency oscillations between the subthalamic nucleus (STN) and the primary motor cortex (M1), are thought to contribute to motor impairment in Parkinson's disease (PD). However, the alterations of electrophysiological characteristics of STN and M1 in PD are still unclear, especially under specific treadmill movement. To examine the relationship between electrophysiological activity in the STN-M1 pathway, extracellular spike trains and local field potential (LFPs) of STN and M1 were simultaneously recorded during resting and movement in unilateral 6-hydroxydopamine (6-OHDA) lesioned rats. The results showed that the identified STN neurons and M1 neurons exhibited abnormal neuronal activity after dopamine loss. The dopamine depletion altered the LFP power in STN and M1 whatever in rest or movement states. Furthermore, the enhanced synchronization of LFP oscillations after dopamine loss was found in 12-35 Hz (beta frequencies) between the STN and M1 during rest and movement. In addition, STN neurons were phase-locked firing to M1 oscillations at 12-35 Hz during rest epochs in 6-OHDA lesioned rats. The dopamine depletion also impaired the anatomical connectivity between the M1 and STN by injecting anterograde neuroanatomical tracing virus into M1 in control and PD rats. Collectively, impairment of' electrophysiological activity and anatomical connectivity in the M1-STN pathway may be the basis for dysfunction of the cortico-basal ganglia circuit, correlating with motor symptoms of PD.
Collapse
Affiliation(s)
- Shuang Sun
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China
| | - Xuenan Wang
- Shandong Institute of Brain Science and Brain-inspired Research, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan 250117, China
| | - Xiaoman Shi
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China
| | - Heyi Fang
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China
| | - Yue Sun
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China
| | - Min Li
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China
| | - Hongyu Han
- Weifang Middle School, Weifang 261031, China
| | - Qin He
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China
| | - Xiaojun Wang
- The First Hospital Affiliated with Shandong First Medicine University, Jinan 250014, China
| | - Xiao Zhang
- Editorial Department of Journal, Shandong Jianzhu University, Jinan 250014, China
| | - Zhi Wei Zhu
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China
| | - Feiyu Chen
- School of International Education, Qilu University of Technology, Jinan 250014, China.
| | - Min Wang
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, 88# Wenhua Road, Jinan 250014, China.
| |
Collapse
|
6
|
Srinivasan SR. Targeting Circuit Abnormalities in Neurodegenerative Disease. Mol Pharmacol 2023; 103:38-44. [PMID: 36310030 DOI: 10.1124/molpharm.122.000563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 02/03/2023] Open
Abstract
Despite significant improvement in our ability to diagnose both common and rare neurodegenerative diseases and understand their underlying biologic mechanisms, there remains a disproportionate lack of effective treatments, reflecting the complexity of these disorders. Successfully advancing novel treatments for neurodegenerative disorders will require reconsideration of traditional approaches, which to date have focused largely on specific disease proteins or cells of origin. This article proposes reframing these diseases as conditions of dysfunctional circuitry as a complement to ongoing efforts. Specifically reviewed is how aberrant spiking is a common downstream mechanism in numerous neurodegenerative diseases, often driven by dysfunction in specific ion channels. Surgical modification of this electrical activity via deep brain stimulation is already an approved modality for many of these disorders. Therefore, restoring proper electrical activity by targeting these channels pharmacologically represents a viable strategy for intervention, not only for symptomatic management but also as a potential disease-modifying therapy. Such an approach is likely to be a promising route to treating these devastating disorders, either as monotherapy or in conjunction with current drugs. SIGNIFICANCE STATEMENT: Despite extensive research and improved understanding of the biology driving neurodegenerative disease, there has not been a concomitant increase in approved therapies. Accordingly, it is time to shift our perspective and recognize these diseases also as disorders of circuitry to further yield novel drug targets and new interventions. An approach focused on treating dysfunctional circuitry has the potential to reduce or reverse patient symptoms and potentially modify disease course.
Collapse
|
7
|
Sasserath T, Robertson AL, Mendez R, Hays TT, Smith E, Cooper H, Akanda N, Rumsey JW, Guo X, Farkhondeh A, Pradhan M, Baumgaertel K, Might M, Rodems S, Zheng W, Hickman JJ. An induced pluripotent stem cell-derived NMJ platform for study of the NGLY1-Congenital Disorder of Deglycosylation. ADVANCED THERAPEUTICS 2022; 5:2200009. [PMID: 36589922 PMCID: PMC9798846 DOI: 10.1002/adtp.202200009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Indexed: 01/05/2023]
Abstract
There are many neurological rare diseases where animal models have proven inadequate or do not currently exist. NGLY1 Deficiency, a congenital disorder of deglycosylation, is a rare disease that predominantly affects motor control, especially control of neuromuscular action. In this study, NGLY1-deficient, patient-derived induced pluripotent stem cells (iPSCs) were differentiated into motoneurons (MNs) to identify disease phenotypes analogous to clinical disease pathology with significant deficits apparent in the NGLY1-deficient lines compared to the control. A neuromuscular junction (NMJ) model was developed using patient and wild type (WT) MNs to study functional differences between healthy and diseased NMJs. Reduced axon length, increased and shortened axon branches, MN action potential (AP) bursting and decreased AP firing rate and amplitude were observed in the NGLY1-deficient MNs in monoculture. When transitioned to the NMJ-coculture system, deficits in NMJ number, stability, failure rate, and synchronicity with indirect skeletal muscle (SkM) stimulation were observed. This project establishes a phenotypic NGLY1 model for investigation of possible therapeutics and investigations into mechanistic deficits in the system.
Collapse
Affiliation(s)
- Trevor Sasserath
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826 USA
| | - Ashley L Robertson
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826 USA
| | - Roxana Mendez
- University of Central Florida, NanoScience Technology Center, 12424 Research Parkway, Suite 400, Orlando, FL 32826 USA
| | - Tristan T Hays
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826 USA
| | - Ethan Smith
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826 USA
| | - Helena Cooper
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826 USA
| | - Nesar Akanda
- University of Central Florida, NanoScience Technology Center, 12424 Research Parkway, Suite 400, Orlando, FL 32826 USA
| | - John W Rumsey
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826 USA
| | - Xiufang Guo
- University of Central Florida, NanoScience Technology Center, 12424 Research Parkway, Suite 400, Orlando, FL 32826 USA
| | - Atena Farkhondeh
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Building C, Room 310W Rockville, MD 20850, USA
| | - Manisha Pradhan
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Building C, Room 310W Rockville, MD 20850, USA
| | - Karsten Baumgaertel
- Travere Therapeutics, 3611 Valley Centre Drive, Suite 300, San Diego, CA, USA
| | - Matthew Might
- University of Alabama at Birmingham, Hugh Kaul Precision Medicine Institute, 510 20th St S, Office 858B, Birmingham, AL 35210, USA
| | - Steven Rodems
- Travere Therapeutics, 3611 Valley Centre Drive, Suite 300, San Diego, CA, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Building C, Room 310W Rockville, MD 20850, USA
| | - James J Hickman
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826 USA
- University of Central Florida, NanoScience Technology Center, 12424 Research Parkway, Suite 400, Orlando, FL 32826 USA
| |
Collapse
|
8
|
Cortical network formation based on subthalamic beta bursts in Parkinson's disease. Neuroimage 2022; 263:119619. [PMID: 36087901 DOI: 10.1016/j.neuroimage.2022.119619] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/02/2022] [Accepted: 09/06/2022] [Indexed: 11/21/2022] Open
Abstract
Recent evidence suggests that beta bursts in subthalamic nucleus (STN) play an important role in Parkinsonian pathophysiology. We studied the spatio-temporal relationship between STN beta bursts and cortical activity in 26 Parkinson's disease (PD) patients undergoing deep brain stimulation (DBS) surgery. Postoperatively, we simultaneously recorded STN local field potentials (LFP) from externalized DBS leads and cortical activity using whole-brain magnetoencephalography. Event-related magnetic fields (ERF) were averaged time-locked to STN beta bursts and subjected to source localization. Our results demonstrate that ERF exhibiting activity significantly different from baseline activity were localized within areas functionally related to associative, limbic, and motor systems as well as regions pertinent for visual and language processing. Our data suggest that STN beta bursts are involved in network formation between STN and cortex. This interaction is in line with the idea of parallel processing within the basal ganglia-cortex loop, specifically within the functional subsystems of the STN (i.e., associative, limbic, motor, and the related cortical areas). ERFs within visual and language-related cortical areas indicate involvement of beta bursts in STN-cortex networks beyond the associative, limbic, and motor loops. In sum, our results highlight the involvement of STN beta bursts in the formation of multiple STN - cortex loops in patients with PD.
Collapse
|
9
|
Single-neuron bursts encode pathological oscillations in subcortical nuclei of patients with Parkinson's disease and essential tremor. Proc Natl Acad Sci U S A 2022; 119:e2205881119. [PMID: 36018837 PMCID: PMC9436336 DOI: 10.1073/pnas.2205881119] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Deep brain stimulation procedures offer an invaluable opportunity to study disease through intracranial recordings from awake patients. Here, we address the relationship between single-neuron and aggregate-level (local field potential; LFP) activities in the subthalamic nucleus (STN) and thalamic ventral intermediate nucleus (Vim) of patients with Parkinson's disease (n = 19) and essential tremor (n = 16), respectively. Both disorders have been characterized by pathologically elevated LFP oscillations, as well as an increased tendency for neuronal bursting. Our findings suggest that periodic single-neuron bursts encode both pathophysiological beta (13 to 33 Hz; STN) and tremor (4 to 10 Hz; Vim) LFP oscillations, evidenced by strong time-frequency and phase-coupling relationships between the bursting and LFP signals. Spiking activity occurring outside of bursts had no relationship to the LFP. In STN, bursting activity most commonly preceded the LFP oscillation, suggesting that neuronal bursting generated within STN may give rise to an aggregate-level LFP oscillation. In Vim, LFP oscillations most commonly preceded bursting activity, suggesting that neuronal firing may be entrained by periodic afferent inputs. In both STN and Vim, the phase-coupling relationship between LFP and high-frequency oscillation (HFO) signals closely resembled the relationships between the LFP and single-neuron bursting. This suggests that periodic single-neuron bursting is likely representative of a higher spatial and temporal resolution readout of periodic increases in the amplitude of HFOs, which themselves may be a higher resolution readout of aggregate-level LFP oscillations. Overall, our results may reconcile "rate" and "oscillation" models of Parkinson's disease and shed light on the single-neuron basis and origin of pathophysiological oscillations in movement disorders.
Collapse
|
10
|
Lubejko ST, Graham RD, Livrizzi G, Schaefer R, Banghart MR, Creed MC. The role of endogenous opioid neuropeptides in neurostimulation-driven analgesia. Front Syst Neurosci 2022; 16:1044686. [PMID: 36591324 PMCID: PMC9794630 DOI: 10.3389/fnsys.2022.1044686] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/18/2022] [Indexed: 12/15/2022] Open
Abstract
Due to the prevalence of chronic pain worldwide, there is an urgent need to improve pain management strategies. While opioid drugs have long been used to treat chronic pain, their use is severely limited by adverse effects and abuse liability. Neurostimulation techniques have emerged as a promising option for chronic pain that is refractory to other treatments. While different neurostimulation strategies have been applied to many neural structures implicated in pain processing, there is variability in efficacy between patients, underscoring the need to optimize neurostimulation techniques for use in pain management. This optimization requires a deeper understanding of the mechanisms underlying neurostimulation-induced pain relief. Here, we discuss the most commonly used neurostimulation techniques for treating chronic pain. We present evidence that neurostimulation-induced analgesia is in part driven by the release of endogenous opioids and that this endogenous opioid release is a common endpoint between different methods of neurostimulation. Finally, we introduce technological and clinical innovations that are being explored to optimize neurostimulation techniques for the treatment of pain, including multidisciplinary efforts between neuroscience research and clinical treatment that may refine the efficacy of neurostimulation based on its underlying mechanisms.
Collapse
Affiliation(s)
- Susan T. Lubejko
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Robert D. Graham
- Department of Anesthesiology, Pain Center, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Giulia Livrizzi
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Robert Schaefer
- Department of Anesthesiology, Pain Center, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Matthew R. Banghart
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, United States
- *Correspondence: Matthew R. Banghart,
| | - Meaghan C. Creed
- Department of Anesthesiology, Pain Center, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, United States
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, United States
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, United States
- Meaghan C. Creed,
| |
Collapse
|
11
|
Sure M, Vesper J, Schnitzler A, Florin E. Dopaminergic Modulation of Spectral and Spatial Characteristics of Parkinsonian Subthalamic Nucleus Beta Bursts. Front Neurosci 2021; 15:724334. [PMID: 34867149 PMCID: PMC8636009 DOI: 10.3389/fnins.2021.724334] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
In Parkinson’s disease (PD), subthalamic nucleus (STN) beta burst activity is pathologically elevated. These bursts are reduced by dopamine and deep brain stimulation (DBS). Therefore, these bursts have been tested as a trigger for closed-loop DBS. To provide better targeted parameters for closed-loop stimulation, we investigate the spatial distribution of beta bursts within the STN and if they are specific to a beta sub-band. Local field potentials (LFP) were acquired in the STN of 27 PD patients while resting. Based on the orientation of segmented DBS electrodes, the LFPs were classified as anterior, postero-medial, and postero-lateral. Each recording lasted 30 min with (ON) and without (OFF) dopamine. Bursts were detected in three frequency bands: ±3 Hz around the individual beta peak frequency, low beta band (lBB), and high beta band (hBB). Medication reduced the duration and the number of bursts per minute but not the amplitude of the beta bursts. The burst amplitude was spatially modulated, while the burst duration and rate were frequency dependent. Furthermore, the hBB burst duration was positively correlated with the akinetic-rigid UPDRS III subscore. Overall, these findings on differential dopaminergic modulation of beta burst parameters suggest that hBB burst duration is a promising target for closed-loop stimulation and that burst parameters could guide DBS programming.
Collapse
Affiliation(s)
- Matthias Sure
- Institute of Clinical Neuroscience and Medical Psychology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Jan Vesper
- Department of Functional Neurosurgery and Stereotaxy, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Alfons Schnitzler
- Institute of Clinical Neuroscience and Medical Psychology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany.,Department of Neurology, Center for Movement Disorders and Neuromodulation, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Esther Florin
- Institute of Clinical Neuroscience and Medical Psychology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
12
|
Tai CH. Subthalamic burst firing: A pathophysiological target in Parkinson's disease. Neurosci Biobehav Rev 2021; 132:410-419. [PMID: 34856222 DOI: 10.1016/j.neubiorev.2021.11.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/16/2021] [Accepted: 11/28/2021] [Indexed: 11/27/2022]
Abstract
Understanding the pathophysiological mechanism of Parkinson's disease (PD) in the subthalamic nucleus (STN) has become a critical issue since deep brain stimulation (DBS) in this region has been proven as an effective treatment for this disease. The STN possesses a special ability to switch from the spike to the burst firing mode in response to dopamine deficiency in parkinsonism, and this STN burst is considered an electrophysiological signature of the cortico-basal ganglia circuit in the brains of PD patients. This review focuses on the role of STN burst firing in the pathophysiology of PD and during DBS. Here, we review existing literature on how STN bursts originate and the specific factors affecting their formation; how STN burst firing causes motor symptoms in PD and how interventions can rescue these symptoms. Finally, the similarities and differences between the two electrophysiological hallmarks of PD, STN burst firing and beta-oscillation, are discussed. STN burst firing should be considered as a pathophysiological target in PD during treatment with DBS.
Collapse
Affiliation(s)
- Chun-Hwei Tai
- Department of Neurology, National Taiwan University Hospital, No. 7, Jhongshan South Road, 100225, Taipei, Taiwan.
| |
Collapse
|
13
|
Changes in Excitability Properties of Ventromedial Motor Thalamic Neurons in 6-OHDA Lesioned Mice. eNeuro 2021; 8:ENEURO.0436-20.2021. [PMID: 33509950 PMCID: PMC7920540 DOI: 10.1523/eneuro.0436-20.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 01/19/2021] [Accepted: 01/24/2021] [Indexed: 12/12/2022] Open
Abstract
The activity of basal ganglia input receiving motor thalamus (BGMT) makes a critical impact on motor cortical processing, but modification in BGMT processing with Parkinsonian conditions has not be investigated at the cellular level. Such changes may well be expected because of homeostatic regulation of neural excitability in the presence of altered synaptic drive with dopamine depletion. We addressed this question by comparing BGMT properties in brain slice recordings between control and unilaterally 6-hydroxydopamine hydrochloride (6-OHDA)-treated adult mice. At a minimum of one month after 6-OHDA treatment, BGMT neurons showed a highly significant increase in intrinsic excitability, which was primarily because of a decrease in M-type potassium current. BGMT neurons after 6-OHDA treatment also showed an increase in T-type calcium rebound spikes following hyperpolarizing current steps. Biophysical computer modeling of a thalamic neuron demonstrated that an increase in rebound spiking can also be accounted for by a decrease in the M-type potassium current. Modeling also showed that an increase in sag with hyperpolarizing steps found after 6-OHDA treatment could in part but not fully be accounted for by the decrease in M-type current. These findings support the hypothesis that homeostatic changes in BGMT neural properties following 6-OHDA treatment likely influence the signal processing taking place in the BG thalamocortical network in Parkinson’s disease.
Collapse
|
14
|
Vegas‐Suárez S, Pisanò CA, Requejo C, Bengoetxea H, Lafuente JV, Morari M, Miguelez C, Ugedo L. 6-Hydroxydopamine lesion and levodopa treatment modify the effect of buspirone in the substantia nigra pars reticulata. Br J Pharmacol 2020; 177:3957-3974. [PMID: 32464686 PMCID: PMC7429490 DOI: 10.1111/bph.15145] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND AND PURPOSE l-DOPA-induced dyskinesia (LID) is considered a major complication in the treatment of Parkinson's disease (PD). Buspirone (5-HT1A partial agonist) have shown promising results in the treatment of PD and LID, however no 5-HT-based treatment has been approved in PD. The present study was aimed to investigate how the substantia nigra pars reticulata (SNr) is affected by buspirone and whether it is a good target to study 5-HT antidyskinetic treatments. EXPERIMENTAL APPROACH Buspirone was studied using in vivo single-unit, electrocorticogram, local field potential recordings along with microdialysis and immunohistochemistry in naïve/sham, 6-hydroxydopamine (6-OHDA)-lesioned or 6-OHDA-lesioned and l-DOPA-treated (6-OHDA/l-DOPA) rats. KEY RESULTS Local buspirone inhibited SNr neuron activity in all groups. However, systemic buspirone reduced burst activity in 6-OHDA-lesioned rats (with or without l-DOPA treatment), whereas 8-OH-DPAT, a full 5-HT1A agonist induced larger inhibitory effects in sham animals. Neither buspirone nor 8-OH-DPAT markedly modified the low-frequency oscillatory activity in the SNr or synchronization within the SNr with the cortex. In addition, local perfusion of buspirone increased GABA and glutamate release in the SNr of naïve and 6-OHDA-lesioned rats but no effect in 6-OHDA/l-DOPA rats. In the 6-OHDA/l-DOPA group, increased 5-HT transporter and decreased 5-HT1A receptor expression was found. CONCLUSIONS AND IMPLICATIONS The effects of buspirone in SNr are influenced by dopamine loss and l-DOPA treatment. The present results suggest that the regulation of burst activity of the SNr induced by DA loss may be a good target to test new drugs for the treatment of PD and LID.
Collapse
Affiliation(s)
- Sergio Vegas‐Suárez
- Department of Pharmacology, Faculty of Medicine and NursingUniversity of the Basque Country (UPV/EHU)LeioaSpain
- Autonomic and Movement Disorders Unit, Neurodegenerative DiseasesBiocruces Health Research InstituteBarakaldoBizkaiaSpain
| | - Clarissa Anna Pisanò
- Department of Medical Sciences, Section of PharmacologyUniversity of FerraraFerraraItaly
- Neuroscience Center and National Institute of NeuroscienceUniversity of FerraraFerraraItaly
| | - Catalina Requejo
- LaNCE, Department of NeuroscienceUniversity of the Basque Country (UPV/EHU)LeioaSpain
| | - Harkaitz Bengoetxea
- LaNCE, Department of NeuroscienceUniversity of the Basque Country (UPV/EHU)LeioaSpain
| | - Jose Vicente Lafuente
- LaNCE, Department of NeuroscienceUniversity of the Basque Country (UPV/EHU)LeioaSpain
| | - Michele Morari
- Department of Medical Sciences, Section of PharmacologyUniversity of FerraraFerraraItaly
- Neuroscience Center and National Institute of NeuroscienceUniversity of FerraraFerraraItaly
| | - Cristina Miguelez
- Department of Pharmacology, Faculty of Medicine and NursingUniversity of the Basque Country (UPV/EHU)LeioaSpain
- Autonomic and Movement Disorders Unit, Neurodegenerative DiseasesBiocruces Health Research InstituteBarakaldoBizkaiaSpain
| | - Luisa Ugedo
- Department of Pharmacology, Faculty of Medicine and NursingUniversity of the Basque Country (UPV/EHU)LeioaSpain
- Autonomic and Movement Disorders Unit, Neurodegenerative DiseasesBiocruces Health Research InstituteBarakaldoBizkaiaSpain
| |
Collapse
|
15
|
Gast R, Schmidt H, Knösche TR. A Mean-Field Description of Bursting Dynamics in Spiking Neural Networks with Short-Term Adaptation. Neural Comput 2020; 32:1615-1634. [PMID: 32687770 DOI: 10.1162/neco_a_01300] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bursting plays an important role in neural communication. At the population level, macroscopic bursting has been identified in populations of neurons that do not express intrinsic bursting mechanisms. For the analysis of phase transitions between bursting and non-bursting states, mean-field descriptions of macroscopic bursting behavior are a valuable tool. In this article, we derive mean-field descriptions of populations of spiking neurons and examine whether states of collective bursting behavior can arise from short-term adaptation mechanisms. Specifically, we consider synaptic depression and spike-frequency adaptation in networks of quadratic integrate-and-fire neurons. Analyzing the mean-field model via bifurcation analysis, we find that bursting behavior emerges for both types of short-term adaptation. This bursting behavior can coexist with steady-state behavior, providing a bistable regime that allows for transient switches between synchronized and nonsynchronized states of population dynamics. For all of these findings, we demonstrate a close correspondence between the spiking neural network and the mean-field model. Although the mean-field model has been derived under the assumptions of an infinite population size and all-to-all coupling inside the population, we show that this correspondence holds even for small, sparsely coupled networks. In summary, we provide mechanistic descriptions of phase transitions between bursting and steady-state population dynamics, which play important roles in both healthy neural communication and neurological disorders.
Collapse
Affiliation(s)
- Richard Gast
- Max Planck Institute for Human Cognitive and Brain Sciences, 04103 Leipzig, Germany
| | - Helmut Schmidt
- Max Planck Institute for Human Cognitive and Brain Sciences, 04103 Leipzig, Germany
| | - Thomas R Knösche
- Max Planck Institute for Human Cognitive and Brain Sciences, 04103 Leipzig, Germany, and Institute for Biomedical Engineering and Informatics, TU 98693 Ilmenau, Germany
| |
Collapse
|
16
|
Helgers SOA, Al Krinawe Y, Alam M, Krauss JK, Schwabe K, Hermann EJ, Al-Afif S. Lesion of the Fastigial Nucleus in Juvenile Rats Deteriorates Rat Behavior in Adulthood, Accompanied by Altered Neuronal Activity in the Medial Prefrontal Cortex. Neuroscience 2020; 442:29-40. [PMID: 32621846 DOI: 10.1016/j.neuroscience.2020.06.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 11/16/2022]
Abstract
The cerebellar cognitive affective syndrome may result from various cerebellar injuries. Although it is not exactly known which anatomical structures are involved, the fastigial nucleus has been thought to play a pivotal role according to recent studies. Here we investigate whether bilateral fastigial nucleus lesions in juvenile rats affect cognitive-associative and limbic related functions in adulthood. Furthermore, potential effects on the neuronal activity in the medial prefrontal cortex (mPFC) and local field coherence with the sensorimotor cortex (SMCtx) were evaluated. The fastigial nucleus was lesioned bilaterally by thermocoagulation via stereotaxically inserted electrodes in 23-day old male Sprague Dawley rats. Naïve and sham-lesioned rats (electrodes inserted above the nucleus and no electrical current applied) served as controls. As adults, all groups were tested for cognitive-associative function, social behavior, and anxiety. Thereafter, electrophysiological recordings were obtained under urethane anesthesia. Finally, lesions and recording sites were histologically verified. Spatial learning in a radial maze test and learning in an operant learning paradigm was disturbed in rats with fastigial lesions. Furthermore, in the elevated plus maze anxiety was enhanced, whereas social behavior was not affected. Electrophysiological recordings showed enhanced local field coherence between mPFC and SMCtx across all frequency bands. Impaired cognitive and affective functions together with enhanced coherence between mPFC and SMCtx after bilateral fastigial nucleus lesions indicate that the fastigial nucleus contribute to the development of the cerebellar cognitive affective syndrome and associated motor behavior.
Collapse
Affiliation(s)
- Simeon O A Helgers
- Department of Neurosurgery, Hannover Medical School, Hannover, Lower Saxony, Germany; DFG Cluster of Excellence, Hearing4all, Germany
| | - Yazeed Al Krinawe
- Department of Neurosurgery, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Mesbah Alam
- Department of Neurosurgery, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Joachim K Krauss
- Department of Neurosurgery, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Kerstin Schwabe
- Department of Neurosurgery, Hannover Medical School, Hannover, Lower Saxony, Germany; DFG Cluster of Excellence, Hearing4all, Germany
| | - Elvis J Hermann
- Department of Neurosurgery, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Shadi Al-Afif
- Department of Neurosurgery, Hannover Medical School, Hannover, Lower Saxony, Germany.
| |
Collapse
|
17
|
Bahuguna J, Sahasranamam A, Kumar A. Uncoupling the roles of firing rates and spike bursts in shaping the STN-GPe beta band oscillations. PLoS Comput Biol 2020; 16:e1007748. [PMID: 32226014 PMCID: PMC7145269 DOI: 10.1371/journal.pcbi.1007748] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 04/09/2020] [Accepted: 02/25/2020] [Indexed: 01/15/2023] Open
Abstract
The excess of 15-30 Hz (β-band) oscillations in the basal ganglia is one of the key signatures of Parkinson's disease (PD). The STN-GPe network is integral to generation and modulation of β band oscillations in basal ganglia. However, the role of changes in the firing rates and spike bursting of STN and GPe neurons in shaping these oscillations has remained unclear. In order to uncouple their effects, we studied the dynamics of STN-GPe network using numerical simulations. In particular, we used a neuron model, in which firing rates and spike bursting can be independently controlled. Using this model, we found that while STN firing rate is predictive of oscillations, GPe firing rate is not. The effect of spike bursting in STN and GPe neurons was state-dependent. That is, only when the network was operating in a state close to the border of oscillatory and non-oscillatory regimes, spike bursting had a qualitative effect on the β band oscillations. In these network states, an increase in GPe bursting enhanced the oscillations whereas an equivalent proportion of spike bursting in STN suppressed the oscillations. These results provide new insights into the mechanisms underlying the transient β bursts and how duration and power of β band oscillations may be controlled by an interplay of GPe and STN firing rates and spike bursts.
Collapse
Affiliation(s)
- Jyotika Bahuguna
- Aix Marseille University, Institute for Systems Neuroscience, Marseille, France
- * E-mail: (JB); (AK)
| | | | - Arvind Kumar
- Department of Computational Science and Technology, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, Stockholm, Sweden
- * E-mail: (JB); (AK)
| |
Collapse
|
18
|
Darbin O, Hatanaka N, Takara S, Kaneko N, Chiken S, Naritoku D, Martino A, Nambu A. Parkinsonism Differently Affects the Single Neuronal Activity in the Primary and Supplementary Motor Areas in Monkeys: An Investigation in Linear and Nonlinear Domains. Int J Neural Syst 2020; 30:2050010. [PMID: 32019380 DOI: 10.1142/s0129065720500100] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The changes in neuronal firing activity in the primary motor cortex (M1) and supplementary motor area (SMA) were compared in monkeys rendered parkinsonian by treatment with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. The neuronal dynamic was characterized using mathematical tools defined in different frameworks (rate, oscillations or complex patterns). Then, and for each cortical area, multivariate and discriminate analyses were further performed on these features to identify those important to differentiate between the normal and the pathological neuronal activity. Our results show a different order in the importance of the features to discriminate the pathological state in each cortical area which suggests that the M1 and the SMA exhibit dissimilarities in their neuronal alterations induced by parkinsonism. Our findings highlight the need for multiple mathematical frameworks to best characterize the pathological neuronal activity related to parkinsonism. Future translational studies are warranted to investigate the causal relationships between cortical region-specificities, dominant pathological hallmarks and symptoms.
Collapse
Affiliation(s)
- Olivier Darbin
- Department of Neurology, University South Alabama, 307 University Blvd, Mobile, AL 36688, USA
| | - Nobuhiko Hatanaka
- Division of System Neurophysiology, National Institute for Physiological Sciences and Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Sayuki Takara
- Division of System Neurophysiology, National Institute for Physiological Sciences and Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Nobuya Kaneko
- Division of System Neurophysiology, National Institute for Physiological Sciences and Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Satomi Chiken
- Division of System Neurophysiology, National Institute for Physiological Sciences and Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Dean Naritoku
- Department of Neurology, University South Alabama, 307 University Blvd, Mobile, AL 36688, USA
| | - Anthony Martino
- Department of Neurology, University South Alabama, 307 University Blvd, Mobile, AL 36688, USA
| | - Atsushi Nambu
- Division of System Neurophysiology, National Institute for Physiological Sciences and Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| |
Collapse
|
19
|
Ririe DG, Boada MD, MacGregor MK, Martin SJ, Strassburg TJ, Kim SA, Eisenach JC, Martin TJ. Incisional Nociceptive Input Impairs Attention-related Behavior and Is Associated with Reduced Neuronal Activity in the Prefrontal Cortex in Rats. Anesthesiology 2019; 129:778-790. [PMID: 29952818 DOI: 10.1097/aln.0000000000002325] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
WHAT WE ALREADY KNOW ABOUT THIS TOPIC WHAT THIS ARTICLE TELLS US THAT IS NEW: BACKGROUND:: Cognitive capacity may be reduced from inflammation, surgery, anesthesia, and pain. In this study, we hypothesized that incision-induced nociceptive input impairs attentional performance and alters neuronal activity in the prefrontal cortex. METHODS Attentional performance was measured in rats by using the titration variant of the 5-choice serial reaction time to determine the effect of surgical incision and anesthesia in a visual attention task. Neuronal activity (single spike and local field potentials) was measured in the medial prefrontal cortex in animals during the task. RESULTS Incision significantly impaired attention postoperatively (area under curve of median cue duration-time 97.2 ± 56.8 [n = 9] vs. anesthesia control 25.5 ± 14.5 s-days [n = 9], P = 0.002; effect size, η = 0.456). Morphine (1 mg/kg) reduced impairment after incision (area under curve of median cue duration-time 31.6 ± 36.7 [n = 11] vs. saline 110 ± 64.7 s-days [n = 10], P < 0.001; η = 0.378). Incision also decreased cell activity (n = 24; 1.48 ± 0.58 vs. control, 2.93 ± 2.02 bursts/min; P = 0.002; η = 0.098) and local field potentials (n = 28; η = 0.111) in the medial prefrontal cortex. CONCLUSIONS These results show that acute postoperative nociceptive input from incision reduces attention-related task performance and decreases neuronal activity in the medial prefrontal cortex. Decreased neuronal activity suggests nociceptive input is more than just a distraction because neuronal activity increases during audiovisual distraction with similar behavioral impairment. This suggests that nociceptive input and the medial prefrontal cortex may contribute to attentional impairment and mild cognitive dysfunction postoperatively. In this regard, pain may affect postoperative recovery and return to normal activities through attentional impairment by contributing to lapses in concentration for routine and complex tasks.
Collapse
Affiliation(s)
- Douglas G Ririe
- From the Pain Mechanisms Lab, Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Jiang X, Yan Y, Wang K, Wei J, Su W, Jia J. Brain state-dependent alterations of corticostriatal synchronized oscillations in awake and anesthetized parkinsonian rats. Brain Res 2019; 1717:214-227. [DOI: 10.1016/j.brainres.2019.04.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 12/29/2022]
|
21
|
Snowball A, Chabrol E, Wykes RC, Shekh-Ahmad T, Cornford JH, Lieb A, Hughes MP, Massaro G, Rahim AA, Hashemi KS, Kullmann DM, Walker MC, Schorge S. Epilepsy Gene Therapy Using an Engineered Potassium Channel. J Neurosci 2019; 39:3159-3169. [PMID: 30755487 PMCID: PMC6468110 DOI: 10.1523/jneurosci.1143-18.2019] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 12/24/2018] [Accepted: 01/21/2019] [Indexed: 12/21/2022] Open
Abstract
Refractory focal epilepsy is a devastating disease for which there is frequently no effective treatment. Gene therapy represents a promising alternative, but treating epilepsy in this way involves irreversible changes to brain tissue, so vector design must be carefully optimized to guarantee safety without compromising efficacy. We set out to develop an epilepsy gene therapy vector optimized for clinical translation. The gene encoding the voltage-gated potassium channel Kv1.1, KCNA1, was codon optimized for human expression and mutated to accelerate the recovery of the channels from inactivation. For improved safety, this engineered potassium channel (EKC) gene was packaged into a nonintegrating lentiviral vector under the control of a cell type-specific CAMK2A promoter. In a blinded, randomized, placebo-controlled preclinical trial, the EKC lentivector robustly reduced seizure frequency in a male rat model of focal neocortical epilepsy characterized by discrete spontaneous seizures. When packaged into an adeno-associated viral vector (AAV2/9), the EKC gene was also effective at suppressing seizures in a male rat model of temporal lobe epilepsy. This demonstration of efficacy in a clinically relevant setting, combined with the improved safety conferred by cell type-specific expression and integration-deficient delivery, identify EKC gene therapy as being ready for clinical translation in the treatment of refractory focal epilepsy.SIGNIFICANCE STATEMENT Pharmacoresistant epilepsy affects up to 0.3% of the population. Although epilepsy surgery can be effective, it is limited by risks to normal brain function. We have developed a gene therapy that builds on a mechanistic understanding of altered neuronal and circuit excitability in cortical epilepsy. The potassium channel gene KCNA1 was mutated to bypass post-transcriptional editing and was packaged in a nonintegrating lentivector to reduce the risk of insertional mutagenesis. A randomized, blinded preclinical study demonstrated therapeutic effectiveness in a rodent model of focal neocortical epilepsy. Adeno-associated viral delivery of the channel to both hippocampi was also effective in a model of temporal lobe epilepsy. These results support clinical translation to address a major unmet need.
Collapse
Affiliation(s)
- Albert Snowball
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
| | - Elodie Chabrol
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
| | - Robert C Wykes
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
| | - Tawfeeq Shekh-Ahmad
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
| | - Jonathan H Cornford
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
| | - Andreas Lieb
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
| | - Michael P Hughes
- UCL School of Pharmacy, University College London, London WC1N 1AX, United Kingdom, and
| | - Giulia Massaro
- UCL School of Pharmacy, University College London, London WC1N 1AX, United Kingdom, and
| | - Ahad A Rahim
- UCL School of Pharmacy, University College London, London WC1N 1AX, United Kingdom, and
| | - Kevan S Hashemi
- Open Source Instruments Inc., Watertown, Massachusetts 02472
| | - Dimitri M Kullmann
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, United Kingdom,
| | - Matthew C Walker
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, United Kingdom,
| | - Stephanie Schorge
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
| |
Collapse
|
22
|
Madar AD, Ewell LA, Jones MV. Temporal pattern separation in hippocampal neurons through multiplexed neural codes. PLoS Comput Biol 2019; 15:e1006932. [PMID: 31009459 PMCID: PMC6476466 DOI: 10.1371/journal.pcbi.1006932] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 03/06/2019] [Indexed: 12/18/2022] Open
Abstract
Pattern separation is a central concept in current theories of episodic memory: this computation is thought to support our ability to avoid confusion between similar memories by transforming similar cortical input patterns of neural activity into dissimilar output patterns before their long-term storage in the hippocampus. Because there are many ways one can define patterns of neuronal activity and the similarity between them, pattern separation could in theory be achieved through multiple coding strategies. Using our recently developed assay that evaluates pattern separation in isolated tissue by controlling and recording the input and output spike trains of single hippocampal neurons, we explored neural codes through which pattern separation is performed by systematic testing of different similarity metrics and various time resolutions. We discovered that granule cells, the projection neurons of the dentate gyrus, can exhibit both pattern separation and its opposite computation, pattern convergence, depending on the neural code considered and the statistical structure of the input patterns. Pattern separation is favored when inputs are highly similar, and is achieved through spike time reorganization at short time scales (< 100 ms) as well as through variations in firing rate and burstiness at longer time scales. These multiplexed forms of pattern separation are network phenomena, notably controlled by GABAergic inhibition, that involve many celltypes with input-output transformations that participate in pattern separation to different extents and with complementary neural codes: a rate code for dentate fast-spiking interneurons, a burstiness code for hilar mossy cells and a synchrony code at long time scales for CA3 pyramidal cells. Therefore, the isolated hippocampal circuit itself is capable of performing temporal pattern separation using multiplexed coding strategies that might be essential to optimally disambiguate multimodal mnemonic representations.
Collapse
Affiliation(s)
- Antoine D. Madar
- Department of Neuroscience, University of Wisconsin-Madison, WI, United States of America
- Neuroscience Training Program, University of Wisconsin-Madison, WI, United States of America
- Department of Neurobiology, Grossman Institute for Neuroscience, Quantitative Biology and Human Behavior, University of Chicago, IL, United States of America
| | - Laura A. Ewell
- Department of Neuroscience, University of Wisconsin-Madison, WI, United States of America
- Institute of Experimental Epileptology and Cognition Research, University of Bonn–Medical Center, Germany
| | - Mathew V. Jones
- Department of Neuroscience, University of Wisconsin-Madison, WI, United States of America
| |
Collapse
|
23
|
Abstract
'Bursting', defined as periods of high-frequency firing of a neuron separated by periods of quiescence, has been observed in various neuronal systems, both in vitro and in vivo. It has been associated with a range of neuronal processes, including efficient information transfer and the formation of functional networks during development, and has been shown to be sensitive to genetic and pharmacological manipulations. Accurate detection of periods of bursting activity is thus an important aspect of characterising both spontaneous and evoked neuronal network activity. A wide variety of computational methods have been developed to detect periods of bursting in spike trains recorded from neuronal networks. In this chapter, we review several of the most popular and successful of these methods.
Collapse
|
24
|
Long-Lasting Electrophysiological After-Effects of High-Frequency Stimulation in the Globus Pallidus: Human and Rodent Slice Studies. J Neurosci 2018; 38:10734-10746. [PMID: 30373767 DOI: 10.1523/jneurosci.0785-18.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 09/28/2018] [Accepted: 10/24/2018] [Indexed: 11/21/2022] Open
Abstract
Deep-brain stimulation (DBS) of the globus pallidus pars interna (GPi) is a highly effective therapy for movement disorders, yet its mechanism of action remains controversial. Inhibition of local neurons because of release of GABA from afferents to the GPi is a proposed mechanism in patients. Yet, high-frequency stimulation (HFS) produces prolonged membrane depolarization mediated by cholinergic neurotransmission in endopeduncular nucleus (EP, GPi equivalent in rodent) neurons. We applied HFS while recording neuronal firing from an adjacent electrode during microelectrode mapping of GPi in awake patients (both male and female) with Parkinson disease (PD) and dystonia. Aside from after-suppression and no change in neuronal firing, high-frequency microstimulation induced after-facilitation in 38% (26/69) of GPi neurons. In neurons displaying after-facilitation, 10 s HFS led to an immediate decrease of bursting in PD, but not dystonia patients. Moreover, the changes of bursting patterns in neurons with after-suppression or no change after HFS, were similar in both patient groups. To explore the mechanisms responsible, we applied HFS in EP brain slices from rats of either sex. As in humans, HFS in EP induced two subtypes of after-excitation: excitation or excitation with late inhibition. Pharmacological experiments determined that the excitation subtype, induced by lower charge density, was dependent on glutamatergic transmission. HFS with higher charge density induced excitation with late inhibition, which involved cholinergic modulation. Therefore HFS with different charge density may affect the local neurons through multiple synaptic mechanisms. The cholinergic system plays a role in mediating the after-facilitatory effects in GPi neurons, and because of their modulatory nature, may provide a basis for both the immediate and delayed effects of GPi-DBS. We propose a new model to explain the mechanisms of DBS in GPi.SIGNIFICANCE STATEMENT Deep-brain stimulation (DBS) in the globus pallidus pars interna (GPi) improves Parkinson disease (PD) and dystonia, yet its mechanisms in GPi remain controversial. Inhibition has been previously described and thought to indicate activation of GABAergic synaptic terminals, which dominate in GPi. Here we report that 10 s high-frequency microstimulation induced after-facilitation of neural firing in a substantial proportion of GPi neurons in humans. The neurons with after-facilitation, also immediately reduced their bursting activities after high-frequency stimulation in PD, but not dystonia patients. Based on these data and further animal experiments, a mechanistic hypothesis involving glutamatergic, GABAergic, and cholinergic synaptic transmission is proposed to explain both short- and longer-term therapeutic effects of DBS in GPi.
Collapse
|
25
|
Spike discharge characteristic of the caudal mesencephalic reticular formation and pedunculopontine nucleus in MPTP-induced primate model of Parkinson disease. Neurobiol Dis 2018; 128:40-48. [PMID: 30086388 DOI: 10.1016/j.nbd.2018.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 07/24/2018] [Accepted: 08/03/2018] [Indexed: 12/20/2022] Open
Abstract
The pedunculopontine nucleus (PPN) included in the caudal mesencephalic reticular formation (cMRF) plays a key role in the control of locomotion and wake state. Regarding its involvement in the neurodegenerative process observed in Parkinson disease (PD), deep brain stimulation of the PPN was proposed to treat levodopa-resistant gait disorders. However, the precise role of the cMRF in the pathophysiology of PD, particularly in freezing of gait and other non-motor symptoms is still not clear. Here, using micro electrode recording (MER) in 2 primates, we show that dopamine depletion did not alter the mean firing rate of the overall cMRF neurons, particularly the putative non-cholinergic ones, but only a decreased activity of the regular neurons sub-group (though to be the cholinergic PPN neurons). Interestingly, a significant increase in the relative proportion of cMRF neurons with a burst pattern discharge was observed after MPTP intoxication. The present results question the hypothesis of an over-inhibition of the CMRF by the basal ganglia output structures in PD. The decreased activity observed in the regular neurons could explain some non-motor symptoms in PD regarding the strong involvement of the cholinergic neurons on the modulation of the thalamo-cortical system. The increased burst activity under dopamine depletion confirms that this specific spike discharge pattern activity also observed in other basal ganglia nuclei and in different pathologies could play a mojor role in the pathophysiology of the disease and could explain several symptoms of PD including the freezing of gait. The present data will have to be replicated in a larger number of animals and will have to investigate more in details how the modification of the spike discharge of the cMRF neurons in the parkinsonian state could alter functions such as locomotion and attentional state. This will ultimely allow a better comprehension of the pathophysiology of freezing of gait.
Collapse
|
26
|
Assini R, Abercrombie ED. Zolpidem ameliorates motor impairments in the unilaterally 6-hydroxydopamine-lesioned rat. Eur J Neurosci 2018; 48:1896-1905. [PMID: 30019535 DOI: 10.1111/ejn.14075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/29/2018] [Accepted: 07/14/2018] [Indexed: 11/29/2022]
Abstract
Nuclei within the basal ganglia-such as the globus pallidus external segment, subthalamic nucleus, and substantia nigra pars reticulata-have been shown to exhibit synchronous bursting activity entrained to excessive cortical beta oscillations following dopamine depletion. Zolpidem binds to GABAA receptors with selectivity for those expressing the α1 subunit, potentiating inhibitory postsynaptic currents and increasing the time decay of channel opening. Interestingly, zolpidem-sensitive nuclei within the basal ganglia circuitry are also those that have been shown to exhibit hyperexcitation in a dopamine-depleted state. We hypothesized that a drug with selectivity for these nuclei may improve motor impairments associated with Parkinson's disease. In order to determine the threshold dose at which zolpidem might encumber motor behavior, a dose-response experiment was performed in intact rats using rotarod. Next, we tested whether subthreshold doses (0.1, 0.25, 0.5 mg/kg; i.p.) of zolpidem improved volitional motor behavior/coordination using the rotarod balance beam and cylinder/paw preference tests in unilaterally 6-hydroxydopamine-lesioned rats. It was found that 0.1 mg/kg zolpidem significantly improved rotarod performance and significantly reduced forelimb use asymmetry compared to undrugged post-lesion conditions. Here, we present the first translational evidence for a role of zolpidem-sensitive GABAA receptors in the treatment of PD motor symptoms. Our data show that zolpidem improves both motor coordination and volitional forelimb use in the unilateral 6-hydroxydopamine lesion model of PD, and thus suggest that zolpidem-sensitive GABAA receptors may represent a novel therapeutic target for the treatment of motor symptoms of Parkinson's disease.
Collapse
Affiliation(s)
- Robert Assini
- Center for Molecular & Behavioral Neuroscience, Rutgers, The State University of New Jersey, Newark, New Jersey
| | - Elizabeth D Abercrombie
- Center for Molecular & Behavioral Neuroscience, Rutgers, The State University of New Jersey, Newark, New Jersey
| |
Collapse
|
27
|
Lin SH, Lai HY, Lo YC, Chou C, Chou YT, Yang SH, Sun I, Chen BW, Wang CF, Liu GT, Jaw FS, Chen SY, Chen YY. Decreased Power but Preserved Bursting Features of Subthalamic Neuronal Signals in Advanced Parkinson's Patients under Controlled Desflurane Inhalation Anesthesia. Front Neurosci 2017; 11:701. [PMID: 29311782 PMCID: PMC5733027 DOI: 10.3389/fnins.2017.00701] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 11/28/2017] [Indexed: 11/13/2022] Open
Abstract
Deep brain stimulation (DBS) surgery of the subthalamic nucleus (STN) under general anesthesia (GA) had been used in Parkinson's disease (PD) patients who are unable tolerate awake surgery. The effect of anesthetics on intraoperative microelectrode recording (MER) remains unclear. Understanding the effect of anesthetics on MER is important in performing STN DBS surgery with general anesthesia. In this study, we retrospectively performed qualitive and quantitative analysis of STN MER in PD patients received STN DBS with controlled desflurane anesthesia or LA and compared their clinical outcome. From January 2005 to March 2006, 19 consecutive PD patients received bilateral STN DBS surgery in Hualien Tzu-Chi hospital under either desflurane GA (n = 10) or LA (n = 9). We used spike analysis (frequency and modified burst index [MBI]) and the Hilbert transform to obtain signal power measurements for background and spikes, and compared the characterizations of intraoperative microelectrode signals between the two groups. Additionally, STN firing pattern characteristics were determined using a combined approach based on the autocorrelogram and power spectral analysis, which was employed to investigate differences in the oscillatory activities between the groups. Clinical outcomes were assessed using the Unified Parkinson's Disease Rating Scale (UPDRS) before and after surgery. The results revealed burst firing was observed in both groups. The firing frequencies were greater in the LA group and MBI was comparable in both groups. Both the background and spikes were of significantly greater power in the LA group. The power spectra of the autocorrelograms were significantly higher in the GA group between 4 and 8 Hz. Clinical outcomes based on the UPDRS were comparable in both groups before and after DBS surgery. Under controlled light desflurane GA, burst features of the neuronal firing patterns are preserved in the STN, but power is reduced. Enhanced low-frequency (4–8 Hz) oscillations in the MERs for the GA group could be a characteristic signature of desflurane's effect on neurons in the STN.
Collapse
Affiliation(s)
- Sheng-Huang Lin
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan.,Department of Neurology, Tzu Chi General Hospital, Tzu Chi University, Hualien, Taiwan
| | - Hsin-Yi Lai
- Interdisciplinary Institute of Neuroscience and Technology, Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China
| | - Yu-Chun Lo
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chin Chou
- Department of Biomedical Engineering, National Yang Ming University, Taipei, Taiwan
| | - Yi-Ting Chou
- Department of Biomedical Engineering, National Yang Ming University, Taipei, Taiwan
| | - Shih-Hung Yang
- Department of Mechanical and Computer Aided Engineering, Feng Chia University, Taichung, Taiwan
| | - I Sun
- Department of Life Sciences, Institute of Genome Sciences, National Yang Ming University, Taipei, China
| | - Bo-Wei Chen
- Department of Biomedical Engineering, National Yang Ming University, Taipei, Taiwan
| | - Ching-Fu Wang
- Department of Biomedical Engineering, National Yang Ming University, Taipei, Taiwan
| | - Guan-Tze Liu
- Department of Medicine, National Yang Ming University, Taipei, Taiwan
| | - Fu-Shan Jaw
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Shin-Yuan Chen
- Department of Neurosurgery, Tzu Chi General Hospital, Tzu Chi University, Hualien, Taiwan
| | - You-Yin Chen
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Department of Biomedical Engineering, National Yang Ming University, Taipei, Taiwan
| |
Collapse
|
28
|
Huang CS, Wang GH, Tai CH, Hu CC, Yang YC. Antiarrhythmics cure brain arrhythmia: The imperativeness of subthalamic ERG K + channels in parkinsonian discharges. SCIENCE ADVANCES 2017; 3:e1602272. [PMID: 28508055 PMCID: PMC5425237 DOI: 10.1126/sciadv.1602272] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 03/15/2017] [Indexed: 06/07/2023]
Abstract
ERG K+ channels have long been known to play a crucial role in shaping cardiac action potentials and, thus, appropriate heart rhythms. The functional role of ERG channels in the central nervous system, however, remains elusive. We demonstrated that ERG channels exist in subthalamic neurons and have similar gating characteristics to those in the heart. ERG channels contribute crucially not only to the setting of membrane potential and, consequently, the firing modes, but also to the configuration of burst discharges and, consequently, the firing frequency and automaticity of the subthalamic neurons. Moreover, modulation of subthalamic discharges via ERG channels effectively modulates locomotor behaviors. ERG channel inhibitors ameliorate parkinsonian symptoms, whereas enhancers render normal animals hypokinetic. Thus, ERG K+ channels could be vital to the regulation of both cardiac and neuronal rhythms and may constitute an important pathophysiological basis and pharmacotherapeutic target for the growing list of neurological disorders related to "brain arrhythmias."
Collapse
Affiliation(s)
- Chen-Syuan Huang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Guan-Hsun Wang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Chun-Hwei Tai
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Chang Hu
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Ya-Chin Yang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Tao-Yuan, Taiwan
| |
Collapse
|
29
|
Ririe DG, Boada MD, Schmidt BS, Martin SJ, Kim SA, Martin TJ. Audiovisual Distraction Increases Prefrontal Cortical Neuronal Activity and Impairs Attentional Performance in the Rat. J Exp Neurosci 2017; 11:1179069517703080. [PMID: 28469479 PMCID: PMC5398228 DOI: 10.1177/1179069517703080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 03/02/2017] [Indexed: 11/16/2022] Open
Abstract
Involvement of attentional processes is generally evidenced by disruption of behavior in the presence of distracting stimuli. The medial prefrontal cortex (mPFC) seems to play a role in fine-tuning activity during attentional tasks. A novel titration variant of the 5-choice serial reaction time task (5-choice serial reaction time titration variant [5CTV]) that adjusts task difficulty based on subject performance was used to evaluate the effects of audiovisual distraction (DSTR) on performance and mPFC single spike activity and local field potential (LFP). Attention was impaired in the 5CTV from DSTR, and mPFC spike activity was increased, whereas LFP was reduced. The increased spike activity in the mPFC in conjunction with DSTR suggests that conflicting attentional demands may contribute to the reduced task performance. As both hypo- and hyperactivation of the mPFC may contribute to attentional disruption, further studies using the 5CTV are needed to understand mPFC activity changes in real time during disruption of performance by other types of behavioral or neurobiological manipulations.
Collapse
Affiliation(s)
- Douglas G Ririe
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - M Danilo Boada
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Benjamin S Schmidt
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Salem J Martin
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Susy A Kim
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Thomas J Martin
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
30
|
Nicotine Modifies Corticostriatal Plasticity and Amphetamine Rewarding Behaviors in Mice(1,2,3). eNeuro 2016; 3:eN-NWR-0095-15. [PMID: 26866057 PMCID: PMC4745180 DOI: 10.1523/eneuro.0095-15.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 11/22/2015] [Accepted: 12/28/2015] [Indexed: 12/20/2022] Open
Abstract
Corticostriatal signaling participates in sensitized responses to drugs of abuse, where short-term increases in dopamine availability provoke persistent, yet reversible, changes in glutamate release. Prior studies in mice show that amphetamine withdrawal promotes a chronic presynaptic depression in glutamate release, whereas an amphetamine challenge reverses this depression by potentiating corticostriatal activity in direct pathway medium spiny neurons. This synaptic plasticity promotes corticostriatal activity and locomotor sensitization through upstream changes in the activity of tonically active cholinergic interneurons (ChIs). We used a model of operant drug-taking behaviors, in which mice self-administered amphetamine through an in-dwelling catheter. Mice acquired amphetamine self-administration under fixed and increasing schedules of reinforcement. Following a period of abstinence, we determined whether nicotinic acetylcholine receptors modified drug-seeking behavior and associated alterations in ChI firing and corticostriatal activity. Mice responding to conditioned reinforcement showed reduced ChI and corticostriatal activity ex vivo, which paradoxically increased following an amphetamine challenge. Nicotine, in a concentration that increases Ca2+ influx and desensitizes α4β2*-type nicotinic receptors, reduced amphetamine-seeking behaviors following abstinence and amphetamine-induced locomotor sensitization. Nicotine blocked the depression of ChI firing and corticostriatal activity and the potentiating response to an amphetamine challenge. Together, these results demonstrate that nicotine reduces reward-associated behaviors following repeated amphetamine and modifies the changes in ChIs firing and corticostriatal activity. By returning glutamatergic activity in amphetamine self-administering mice to a more stable and normalized state, nicotine limits the depression of striatal activity in withdrawal and the increase in activity following abstinence and a subsequent drug challenge.
Collapse
|
31
|
Subthalamic nucleus phase-amplitude coupling correlates with motor impairment in Parkinson's disease. Clin Neurophysiol 2016; 127:2010-9. [PMID: 26971483 PMCID: PMC4803022 DOI: 10.1016/j.clinph.2016.01.015] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Revised: 12/18/2015] [Accepted: 01/16/2016] [Indexed: 01/05/2023]
Abstract
We obtained invasive subthalamic nucleus recordings in 33 Parkinson’s disease patients. Phase–amplitude coupling between beta band and high-frequency oscillations correlates with severity of motor impairments. Parkinsonian pathophysiology is more closely linked with low-beta band frequencies.
Objective High-amplitude beta band oscillations within the subthalamic nucleus are frequently associated with Parkinson’s disease but it is unclear how they might lead to motor impairments. Here we investigate a likely pathological coupling between the phase of beta band oscillations and the amplitude of high-frequency oscillations around 300 Hz. Methods We analysed an extensive data set comprising resting-state recordings obtained from deep brain stimulation electrodes in 33 patients before and/or after taking dopaminergic medication. We correlated mean values of spectral power and phase–amplitude coupling with severity of hemibody bradykinesia/rigidity. In addition, we used simultaneously recorded magnetoencephalography to look at functional interactions between the subthalamic nucleus and ipsilateral motor cortex. Results Beta band power and phase–amplitude coupling within the subthalamic nucleus correlated positively with severity of motor impairment. This effect was more pronounced within the low-beta range, whilst coherence between subthalamic nucleus and motor cortex was dominant in the high-beta range. Conclusions We speculate that the beta band might impede pro-kinetic high-frequency activity patterns when phase–amplitude coupling is prominent. Furthermore, results provide evidence for a functional subdivision of the beta band into low and high frequencies. Significance Our findings contribute to the interpretation of oscillatory activity within the cortico-basal ganglia circuit.
Collapse
|
32
|
Maurice N, Liberge M, Jaouen F, Ztaou S, Hanini M, Camon J, Deisseroth K, Amalric M, Kerkerian-Le Goff L, Beurrier C. Striatal Cholinergic Interneurons Control Motor Behavior and Basal Ganglia Function in Experimental Parkinsonism. Cell Rep 2015; 13:657-666. [PMID: 26489458 DOI: 10.1016/j.celrep.2015.09.034] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 08/25/2015] [Accepted: 09/10/2015] [Indexed: 01/29/2023] Open
Abstract
Despite evidence showing that anticholinergic drugs are of clinical relevance in Parkinson's disease (PD), the causal role of striatal cholinergic interneurons (CINs) in PD pathophysiology remains elusive. Here, we show that optogenetic inhibition of CINs alleviates motor deficits in PD mouse models, providing direct demonstration for their implication in parkinsonian motor dysfunctions. As neural correlates, CIN inhibition in parkinsonian mice differentially impacts the excitability of striatal D1 and D2 medium spiny neurons, normalizes pathological bursting activity in the main basal ganglia output structure, and increases the functional weight of the direct striatonigral pathway in cortical information processing. By contrast, CIN inhibition in non-lesioned mice does not affect locomotor activity, equally modulates medium spiny neuron excitability, and does not modify spontaneous or cortically driven activity in the basal ganglia output, suggesting that the role of these interneurons in motor function is highly dependent on dopamine tone.
Collapse
Affiliation(s)
- Nicolas Maurice
- Aix-Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), UMR 7288, Institut de Biologie du Développement de Marseille (IBDM), 13288 Marseille cedex 9, France
| | - Martine Liberge
- Aix-Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), UMR 7291, FR3C 3512, Laboratoire de Neurosciences Cognitives, 13331 Marseille cedex 3, France
| | - Florence Jaouen
- Aix-Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), UMR 7288, Institut de Biologie du Développement de Marseille (IBDM), 13288 Marseille cedex 9, France
| | - Samira Ztaou
- Aix-Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), UMR 7291, FR3C 3512, Laboratoire de Neurosciences Cognitives, 13331 Marseille cedex 3, France
| | - Marwa Hanini
- Aix-Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), UMR 7288, Institut de Biologie du Développement de Marseille (IBDM), 13288 Marseille cedex 9, France
| | - Jeremy Camon
- Aix-Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), UMR 7291, FR3C 3512, Laboratoire de Neurosciences Cognitives, 13331 Marseille cedex 3, France
| | - Karl Deisseroth
- Departments of Bioengineering and Psychiatry and Howard Hughes Medical Institute, Stanford University, Palo Alto, CA 94305, USA
| | - Marianne Amalric
- Aix-Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), UMR 7291, FR3C 3512, Laboratoire de Neurosciences Cognitives, 13331 Marseille cedex 3, France
| | - Lydia Kerkerian-Le Goff
- Aix-Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), UMR 7288, Institut de Biologie du Développement de Marseille (IBDM), 13288 Marseille cedex 9, France
| | - Corinne Beurrier
- Aix-Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), UMR 7288, Institut de Biologie du Développement de Marseille (IBDM), 13288 Marseille cedex 9, France.
| |
Collapse
|
33
|
Abstract
Neurostimulation as a therapeutic tool has been developed and used for a range of different diseases such as Parkinson's disease, epilepsy, and migraine. However, it is not known why the efficacy of the stimulation varies dramatically across patients or why some patients suffer from severe side effects. This is largely due to the lack of mechanistic understanding of neurostimulation. Hence, theoretical computational approaches to address this issue are in demand. This chapter provides a review of mechanistic computational modeling of brain stimulation. In particular, we will focus on brain diseases, where mechanistic models (e.g., neural population models or detailed neuronal models) have been used to bridge the gap between cellular-level processes of affected neural circuits and the symptomatic expression of disease dynamics. We show how such models have been, and can be, used to investigate the effects of neurostimulation in the diseased brain. We argue that these models are crucial for the mechanistic understanding of the effect of stimulation, allowing for a rational design of stimulation protocols. Based on mechanistic models, we argue that the development of closed-loop stimulation is essential in order to avoid inference with healthy ongoing brain activity. Furthermore, patient-specific data, such as neuroanatomic information and connectivity profiles obtainable from neuroimaging, can be readily incorporated to address the clinical issue of variability in efficacy between subjects. We conclude that mechanistic computational models can and should play a key role in the rational design of effective, fully integrated, patient-specific therapeutic brain stimulation.
Collapse
|
34
|
Lobb CJ, Jaeger D. Bursting activity of substantia nigra pars reticulata neurons in mouse parkinsonism in awake and anesthetized states. Neurobiol Dis 2015; 75:177-85. [PMID: 25576395 DOI: 10.1016/j.nbd.2014.12.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 12/20/2014] [Accepted: 12/24/2014] [Indexed: 01/24/2023] Open
Abstract
Electrophysiological changes in basal ganglia neurons are hypothesized to underlie motor dysfunction in Parkinson's disease (PD). Previous results in head-restrained MPTP-treated non-human primates have suggested that increased bursting within the basal ganglia and related thalamic and cortical areas may be a hallmark of pathophysiological activity. In this study, we investigated whether there is increased bursting in substantia nigra pars reticulata (SNpr) output neurons in anesthetized and awake, head-restrained unilaterally lesioned 6-OHDA mice when compared to control mice. Confirming previous studies, we show that there are significant changes in the firing rate and pattern in SNpr neuron activity under urethane anesthesia. The regular firing pattern of control urethane-anesthetized SNpr neurons was not present in the 6-OHDA-lesioned group, as the latter neurons instead became phase locked with cortical slow wave activity (SWA). Next, we examined whether such robust electrophysiological changes between groups carried over to the awake state. SNpr neurons from both groups fired at much higher frequencies in the awake state than in the anesthetized state and surprisingly showed only modest changes between awake control and 6-OHDA groups. While there were no differences in firing rate between groups in the awake state, an increase in the coefficient of variation (CV) was observed in the 6-OHDA group. Contrary to the bursting hypothesis, this increased CV was not due to changes in bursting but was instead due to a mild increase in pausing. Together, these results suggest that differences in SNpr activity between control and 6-OHDA lesioned mice may be strongly influenced by changes in network activity during different arousal and behavioral states.
Collapse
Affiliation(s)
- C J Lobb
- Department of Biology, Emory University, Atlanta, GA 30322, USA
| | - D Jaeger
- Department of Biology, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
35
|
Alam M, Angelov S, Stemmler M, von Wrangel C, Krauss JK, Schwabe K. Neuronal activity of the prefrontal cortex is reduced in rats selectively bred for deficient sensorimotor gating. Prog Neuropsychopharmacol Biol Psychiatry 2015; 56:174-84. [PMID: 25220677 DOI: 10.1016/j.pnpbp.2014.08.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 08/08/2014] [Accepted: 08/15/2014] [Indexed: 12/24/2022]
Abstract
Rats selectively bred for deficient prepulse inhibition (PPI), an operant measure of sensorimotor gating in which a weak prepulse stimulus attenuates the response to a subsequent startling stimulus, may be used to study certain pathophysiological mechanisms and therapeutic strategies for neuropsychiatric disorders with abnormalities in information processing, such as schizophrenia and Tourette's syndrome (TS). Little is known about neuronal activity in the medial prefrontal cortex (mPFC) and the nucleus accumbens (NAC), which are involved in the modulation of PPI. Here, we examined neuronal activity in these structures, and also in the entopeduncular nucleus (EPN), since lesions of this region alleviate the PPI deficit. Male rats with breeding-induced high and low expression of PPI (n=7, each) were anesthetized with urethane (1.4 mg/kg). Single-unit activity and local field potentials were recorded in the mPFC, the NAC and in the EPN. In the mPFC discharge rate, measures of irregularity and burst activity were significantly reduced in PPI low compared to PPI high rats (P<0.05), while analysis in the NAC showed approximately inverse behavior. In the EPN no difference between groups was found. Additionally, the oscillatory theta band activity (4-8 Hz) was enhanced and the beta band (13-30 Hz) and gamma band (30-100 Hz) activity was reduced in the NAC in PPI low rats. Reduced neuronal activity in the mPFC and enhanced activity in the NAC of PPI low rats, together with altered oscillatory behavior are clearly associated with reduced PPI. PPI low rats may thus be used to study the pathophysiology and therapeutic strategies for neuropsychiatric disorders accompanied by deficient sensorimotor gating.
Collapse
Affiliation(s)
- Mesbah Alam
- Department of Neurosurgery, Hannover Medical School, Carl-Neuberg-Str.1, D- 30625 Hannover, Germany
| | - Svilen Angelov
- Department of Neurosurgery, Hannover Medical School, Carl-Neuberg-Str.1, D- 30625 Hannover, Germany
| | - Meike Stemmler
- Department of Neurosurgery, Hannover Medical School, Carl-Neuberg-Str.1, D- 30625 Hannover, Germany
| | - Christof von Wrangel
- Department of Neurosurgery, Hannover Medical School, Carl-Neuberg-Str.1, D- 30625 Hannover, Germany
| | - Joachim K Krauss
- Department of Neurosurgery, Hannover Medical School, Carl-Neuberg-Str.1, D- 30625 Hannover, Germany
| | - Kerstin Schwabe
- Department of Neurosurgery, Hannover Medical School, Carl-Neuberg-Str.1, D- 30625 Hannover, Germany.
| |
Collapse
|
36
|
Dong H, Li R, Yu C, Xu T, Zhang X, Dong M. Paeoniflorin inhibition of 6-hydroxydopamine-induced apoptosis in PC12 cells via suppressing reactive oxygen species-mediated PKCδ/NF-κB pathway. Neuroscience 2015; 285:70-80. [DOI: 10.1016/j.neuroscience.2014.11.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Revised: 11/03/2014] [Accepted: 11/05/2014] [Indexed: 01/25/2023]
|