1
|
Peña-Ortega F. Microglial modulation of neuronal network function and plasticity. J Neurophysiol 2025; 133:661-680. [PMID: 39819084 DOI: 10.1152/jn.00458.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/08/2024] [Accepted: 12/30/2024] [Indexed: 01/19/2025] Open
Abstract
Microglia are the resident immune cells of the central nervous system (CNS), which have been classically viewed as involved in CNS responses to damage and tissue repair. However, microglia are constantly sensing neuronal network activity and changes in the CNS milieu, establishing complex state-dependent microglia-neuron interactions that impact their functions. By doing so, microglia perform a wide range of physiological roles, including brain homeostasis maintenance, control of neural connectivity, network function modulation, as well as functional and morphological plasticity regulation in health and disease. Here, the author reviews recent evidence of the modulations induced by microglia, a highly heterogeneous cell type, on synaptic and intrinsic neuronal properties, and on neuronal network patterns during perinatal development and adulthood. The reviewed evidence clearly indicates that microglia are important, if not essential, for brain function and plasticity in both health and disease.
Collapse
Affiliation(s)
- Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Mexico
| |
Collapse
|
2
|
Dalla Porta L, Barbero-Castillo A, Sanchez-Sanchez JM, Sanchez-Vives MV. M-current modulation of cortical slow oscillations: Network dynamics and computational modeling. PLoS Comput Biol 2023; 19:e1011246. [PMID: 37405991 DOI: 10.1371/journal.pcbi.1011246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 06/06/2023] [Indexed: 07/07/2023] Open
Abstract
The slow oscillation is a synchronized network activity expressed by the cortical network in slow wave sleep and under anesthesia. Waking up requires a transition from this synchronized brain state to a desynchronized one. Cholinergic innervation is critical for the transition from slow-wave-sleep to wakefulness, and muscarinic action is largely exerted through the muscarinic-sensitive potassium current (M-current) block. We investigated the dynamical impact of blocking the M-current on slow oscillations, both in cortical slices and in a cortical network computational model. Blocking M-current resulted in an elongation of Up states (by four times) and in a significant firing rate increase, reflecting an increased network excitability, albeit no epileptiform discharges occurred. These effects were replicated in a biophysical cortical model, where a parametric reduction of the M-current resulted in a progressive elongation of Up states and firing rate. All neurons, and not only those modeled with M-current, increased their firing rates due to network recurrency. Further increases in excitability induced even longer Up states, approaching the microarousals described in the transition towards wakefulness. Our results bridge an ionic current with network modulation, providing a mechanistic insight into network dynamics of awakening.
Collapse
Affiliation(s)
- Leonardo Dalla Porta
- Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | | | - Maria V Sanchez-Vives
- Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- ICREA, Passeig Lluís Companys, Barcelona, Spain
| |
Collapse
|
3
|
Mueller SAL, Oler JA, Roseboom PH, Aggarwal N, Kenwood MM, Riedel MK, Elam VR, Olsen ME, DiFilippo AH, Christian BT, Hu X, Galvan A, Boehm MA, Michaelides M, Kalin NH. DREADD-mediated amygdala activation is sufficient to induce anxiety-like responses in young nonhuman primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.06.543911. [PMID: 37333300 PMCID: PMC10274719 DOI: 10.1101/2023.06.06.543911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Anxiety disorders are among the most prevalent psychiatric disorders, with symptoms often beginning early in life. To model the pathophysiology of human pathological anxiety, we utilized Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) in a nonhuman primate model of anxious temperament to selectively increase neuronal activity of the amygdala. Subjects included 10 young rhesus macaques; 5 received bilateral infusions of AAV5-hSyn-HA-hM3Dq into the dorsal amygdala, and 5 served as controls. Subjects underwent behavioral testing in the human intruder paradigm following clozapine or vehicle administration, prior to and following surgery. Behavioral results indicated that clozapine treatment post-surgery increased freezing across different threat-related contexts in hM3Dq subjects. This effect was again observed approximately 1.9 years following surgery, indicating the long-term functional capacity of DREADD-induced neuronal activation. [11C]deschloroclozapine PET imaging demonstrated amygdala hM3Dq-HA specific binding, and immunohistochemistry revealed that hM3Dq-HA expression was most prominent in basolateral nuclei. Electron microscopy confirmed expression was predominantly on neuronal membranes. Together, these data demonstrate that activation of primate amygdala neurons is sufficient to induce increased anxiety-related behaviors, which could serve as a model to investigate pathological anxiety in humans.
Collapse
Affiliation(s)
- Sascha A L Mueller
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Jonathan A Oler
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Patrick H Roseboom
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Nakul Aggarwal
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Margaux M Kenwood
- Department of Psychiatry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Marissa K Riedel
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Victoria R Elam
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Miles E Olsen
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Alexandra H DiFilippo
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Bradley T Christian
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Xing Hu
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Adriana Galvan
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Matthew A Boehm
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Ned H Kalin
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| |
Collapse
|
4
|
Méndez-Salcido FA, Torres-Flores MI, Ordaz B, Peña-Ortega F. Abnormal innate and learned behavior induced by neuron-microglia miscommunication is related to CA3 reconfiguration. Glia 2022; 70:1630-1651. [PMID: 35535571 DOI: 10.1002/glia.24185] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 12/15/2022]
Abstract
Neuron-microglia communication through the Cx3cr1-Cx3cl1 axis is essential for the development and refinement of neural circuits, which determine their function into adulthood. In the present work we set out to extend the behavioral characterization of Cx3cr1-/- mice evaluating innate behaviors and spatial navigation, both dependent on hippocampal function. Our results show that Cx3cr1-deficient mice, which show some changes in microglial and synaptic terminals morphology and density, exhibit alterations in activities of daily living and in the rapid encoding of novel spatial information that, nonetheless, improves with training. A neural substrate for these cognitive deficiencies was found in the form of synaptic dysfunction in the CA3 region of the hippocampus, with a marked impact on the mossy fiber (MF) pathway. A network analysis of the CA3 microcircuit reveals the effect of these synaptic alterations on the functional connectivity among CA3 neurons with diminished strength and topological reorganization in Cx3cr1-deficient mice. Neonatal population activity of the CA3 region in Cx3cr1-deficient mice shows a marked reorganization around the giant depolarizing potentials, the first form of network-driven activity of the hippocampus, suggesting that alterations found in adult subjects arise early on in postnatal development, a critical period of microglia-dependent neural circuit refinement. Our results show that interruption of the Cx3cr1-Cx3cl1/neuron-microglia axis leads to changes in CA3 configuration that affect innate and learned behaviors.
Collapse
Affiliation(s)
- Felipe Antonio Méndez-Salcido
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México
| | - Mayra Itzel Torres-Flores
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México
| | - Benito Ordaz
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México
| |
Collapse
|
5
|
Zhai J, Zhou YY, Lagrutta A. Sensitivity, specificity and limitation of in vitro hippocampal slice and neuron-based assays for assessment of drug-induced seizure liability. Toxicol Appl Pharmacol 2021; 430:115725. [PMID: 34536444 DOI: 10.1016/j.taap.2021.115725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 09/01/2021] [Accepted: 09/13/2021] [Indexed: 10/20/2022]
Abstract
An effective in vitro screening assay to detect seizure liability in preclinical development can contribute to better lead molecule optimization prior to candidate selection, providing higher throughput and overcoming potential brain exposure limitations in animal studies. This study explored effects of 26 positive and 14 negative reference pharmacological agents acting through different mechanisms, including 18 reference agents acting on glutamate signaling pathways, in a brain slice assay (BSA) of adult rat to define the assay's sensitivity, specificity, and limitations. Evoked population spikes (PS) were recorded from CA1 pyramidal neurons of hippocampus (HPC) in the BSA. Endpoints for analysis were PS area and PS number. Most positive references (24/26) elicited a concentration-dependent increase in PS area and/or PS number. The negative references (14/14) had little effect on the PS. Moreover, we studied the effects of 15 reference agents testing positive in the BSA on spontaneous activity in E18 rat HPC neurons monitored with microelectrode arrays (MEA), and compared these effects to the BSA results. From these in vitro studies we conclude that the BSA provides 93% sensitivity and 100% specificity in prediction of drug-induced seizure liability, including detecting seizurogenicity by 3 groups of metabotropic glutamate receptor (mGluR) ligands. The MEA results seemed more variable, both quantitatively and directionally, particularly for endpoints capturing synchronized electrical activity. We discuss these results from the two models, comparing each with published results, and provide potential explanations for differences and future directions.
Collapse
Affiliation(s)
- Jin Zhai
- Department of Genetic Toxicology and In Vitro Cellular Toxicity, Safety Assessment & Laboratory Animal Resources (SALAR), Merck & Co., Inc., West Point, PA 19486, USA.
| | - Ying-Ying Zhou
- Program Discovery and Development, Safety Assessment & Laboratory Animal Resources (SALAR), Merck & Co., Inc., West Point, PA 19486, USA
| | - Armando Lagrutta
- Program Discovery and Development, Safety Assessment & Laboratory Animal Resources (SALAR), Merck & Co., Inc., West Point, PA 19486, USA
| |
Collapse
|
6
|
Naffaa MM, Al-Ewaidat OA. Ligand modulation of KCNQ-encoded (K V7) potassium channels in the heart and nervous system. Eur J Pharmacol 2021; 906:174278. [PMID: 34174270 DOI: 10.1016/j.ejphar.2021.174278] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/06/2021] [Accepted: 06/18/2021] [Indexed: 10/21/2022]
Abstract
KCNQ-encoded (KV7) potassium channels are diversely distributed in the human tissues, associated with many physiological processes and pathophysiological conditions. These channels are increasingly used as drug targets for treating diseases. More selective and potent molecules on various types of the KV7 channels are desirable for appropriate therapies. The recent knowledge of the structure and function of human KCNQ-encoded channels makes it more feasible to achieve these goals. This review discusses the role and mechanism of action of many molecules in modulating the function of the KCNQ-encoded potassium channels in the heart and nervous system. The effects of these compounds on KV7 channels help to understand their involvement in many diseases, and to search for more selective and potent ligands to be used in the treatment of many disorders such as various types of cardiac arrhythmias, epilepsy, and pain.
Collapse
Affiliation(s)
- Moawiah M Naffaa
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA; Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA.
| | - Ola A Al-Ewaidat
- Faculty of Medicine, The University of Jordan, Amman, 11942, Jordan
| |
Collapse
|
7
|
Martínez-García I, Hernández-Soto R, Villasana-Salazar B, Ordaz B, Peña-Ortega F. Alterations in Piriform and Bulbar Activity/Excitability/Coupling Upon Amyloid-β Administration in vivo Related to Olfactory Dysfunction. J Alzheimers Dis 2021; 82:S19-S35. [PMID: 33459655 DOI: 10.3233/jad-201392] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Deficits in odor detection and discrimination are premature symptoms of Alzheimer's disease (AD) that correlate with pathological signs in the olfactory bulb (OB) and piriform cortex (PCx). Similar olfactory dysfunction has been characterized in AD transgenic mice that overproduce amyloid-β peptide (Aβ), which can be prevented by reducing Aβ levels by immunological and pharmacological means, suggesting that olfactory dysfunction depends on Aβ accumulation and Aβ-driven alterations in the OB and/or PCx, as well as on their activation. However, this possibility needs further exploration. OBJECTIVE To characterize the effects of Aβ on OB and PCx excitability/coupling and on olfaction. METHODS Aβ oligomerized solution (containing oligomers, monomers, and protofibrils) or its vehicle were intracerebroventricularlly injected two weeks before OB and PCx excitability and synchrony were evaluated through field recordings in vivo and in brain slices. Synaptic transmission from the OB to the PCx was also evaluated in slices. Olfaction was assessed through the habituation/dishabituation test. RESULTS Aβ did not affect lateral olfactory tract transmission into the PCx but reduced odor habituation and cross-habituation. This olfactory dysfunction was related to a reduction of PCx and OB network activity power in vivo. Moreover, the coherence between PCx-OB activities was also reduced by Aβ. Finally, Aβ treatment exacerbated the 4-aminopyridine-induced excitation in the PCx in slices. CONCLUSION Our results show that Aβ-induced olfactory dysfunction involves a complex set of pathological changes at different levels of the olfactory pathway including alterations in PCx excitability and its coupling with the OB. These pathological changes might contribute to hyposmia in AD.
Collapse
Affiliation(s)
- Ignacio Martínez-García
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, UNAM-Campus Juriquilla, México
| | - Rebeca Hernández-Soto
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, UNAM-Campus Juriquilla, México
| | - Benjamín Villasana-Salazar
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, UNAM-Campus Juriquilla, México
| | - Benito Ordaz
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, UNAM-Campus Juriquilla, México
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, UNAM-Campus Juriquilla, México
| |
Collapse
|
8
|
Hung TY, Huang CW, Wu SN. High ability of zileuton ((±)-1-(1-benzo[b]thien-2-ylethyl)-1-hydroxyurea) to stimulate I K(Ca) but suppress I K(DR) and I K(M) independently of 5-lipoxygenase inhibition. Eur J Pharmacol 2020; 887:173482. [PMID: 32795513 DOI: 10.1016/j.ejphar.2020.173482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 08/07/2020] [Accepted: 08/08/2020] [Indexed: 12/16/2022]
Abstract
Zileuton (Zyflo®) is regarded to be an inhibitor of 5-lipoxygenase. Although its effect on Ca2+-activated K+ currents has been reported, its overall ionic effects on neurons are uncertain. In whole-cell current recordings, zileuton increased the amplitude of Ca2+-activated K+ currents with an EC50 of 3.2 μM in pituitary GH3 lactotrophs. Furthermore, zileuton decreased the amplitudes of both delayed-rectifier K+ current (IK(DR)) and M-type K+ current (IK(M)). Conversely, no modification of hyperpolarization-activated cation current (Ih) was demonstrated in its presence of zileuton, although the subsequent addition of cilobradine effectively suppressed the current. In inside-out current recordings, the addition of zileuton to the bath increased the probability of large-conductance Ca2+-activated K+ (BKCa) channels; however, the subsequent addition of GAL-021 effectively reversed the stimulation of channel activity. The kinetic analyses showed an evident shortening in the slow component of mean closed time of BKCa channels in the presence of zileuton, with minimal change in mean open time or that in the fast component of mean closed time. The elevation of BKCa channels caused by zileuton was also observed in hippocampal mHippoE-14 neurons, without any modification of single-channel amplitude. In conclusion, except for its suppression of 5-lipoxygenase, our results indicate that zileuton does not exclusively act on BKCa channels, and its inhibitory effects on IK(DR) and IK(M) may combine to exert strong influence on the functional activities of electrically excitable cells in vivo.
Collapse
Affiliation(s)
- Te-Yu Hung
- Department of Pediatrics, Chi-Mei Medical Center, Tainan, Taiwan
| | - Chin-Wei Huang
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan, Taiwan; Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung City, Taiwan.
| |
Collapse
|
9
|
Villasana-Salazar B, Hernández-Soto R, Guerrero-Gómez ME, Ordaz B, Manrique-Maldonado G, Salgado-Puga K, Peña-Ortega F. Chronic intermittent hypoxia transiently increases hippocampal network activity in the gamma frequency band and 4-Aminopyridine-induced hyperexcitability in vitro. Epilepsy Res 2020; 166:106375. [PMID: 32745888 DOI: 10.1016/j.eplepsyres.2020.106375] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/21/2020] [Accepted: 05/28/2020] [Indexed: 12/12/2022]
Abstract
Chronic intermittent hypoxia (CIH) is the most distinct feature of obstructive sleep apnea (OSA), a common breathing and sleep disorder that leads to several neuropathological consequences, including alterations in the hippocampal network and in seizure susceptibility. However, it is currently unknown whether these alterations are permanent or remit upon normal oxygenation. Here, we investigated the effects of CIH on hippocampal spontaneous network activity and hyperexcitability in vitro and explored whether these alterations endure or fade after normal oxygenation. Results showed that applying CIH for 21 days to adult rats increases gamma-band hippocampal network activity and aggravates 4-Aminopyridine-induced epileptiform activity in vitro. Interestingly, these CIH-induced alterations remit after 30 days of normal oxygenation. Our findings indicate that hippocampal network alterations and increased seizure susceptibility induced by CIH are not permanent and can be spontaneously reverted, suggesting that therapeutic interventions against OSA in patients with epilepsy, such as surgery or continuous positive airway pressure (CPAP), could be favorable for seizure control.
Collapse
Affiliation(s)
- Benjamín Villasana-Salazar
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO 76230, México
| | - Rebeca Hernández-Soto
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO 76230, México
| | - María Estefanía Guerrero-Gómez
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO 76230, México
| | - Benito Ordaz
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO 76230, México
| | - Guadalupe Manrique-Maldonado
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO 76230, México
| | - Karla Salgado-Puga
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO 76230, México
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO 76230, México.
| |
Collapse
|
10
|
Lu TL, Gao ZH, Li SW, Wu SN. High Efficacy by GAL-021: A Known Intravenous Peripheral Chemoreceptor Modulator that Suppresses BK Ca-Channel Activity and Inhibits IK(M) or Ih. Biomolecules 2020; 10:188. [PMID: 31991782 PMCID: PMC7072225 DOI: 10.3390/biom10020188] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/25/2022] Open
Abstract
: GAL-021 has recently been developed as a novel breathing control modulator. However, modifications of ionic currents produced by this agent remain uncertain, although its efficacy in suppressing the activity of big-conductance Ca2+-activated K+ (BKCa) channels has been reported. In pituitary tumor (GH3) cells, we found that the presence of GAL-021 decreased the amplitude of macroscopic Ca2+-activated K+ current (IK(Ca)) in a concentration-dependent manner with an effective IC50 of 2.33 μM. GAL-021-mediated reduction of IK(Ca) was reversed by subsequent application of verteporfin or ionomycin; however, it was not by that of diazoxide. In inside-out current recordings, the addition of GAL-021 to the bath markedly decreased the open-state probability of BKCa channels. This agent also resulted in a rightward shift in voltage dependence of the activation curve of BKCa channels; however, neither the gating charge of the curve nor single-channel conductance of the channel was changed. There was an evident lengthening of the mean closed time of BKCa channels in the presence of GAL-021, with no change in mean open time. The GAL-021 addition also suppressed M-type K+ current with an effective IC50 of 3.75 μM; however, its presence did not alter the amplitude of erg-mediated K+ current, or mildly suppressed delayed-rectifier K+ current. GAL-021 at a concentration of 30 μM could also suppress hyperpolarization-activated cationic current. In HEK293T cells expressing α-hSlo, the addition of GAL-021 was also able to suppress the BKCa-channel open probabilities, and GAL-021-mediated suppression of BKCa-channel activity was attenuated by further addition of BMS-191011. Collectively, the GAL-021 effects presented herein do not exclusively act on BKCa channels and these modifications on ionic currents exert significant influence on the functional activities of electrically excitable cells occurring in vivo.
Collapse
Affiliation(s)
- Te-Ling Lu
- School of Pharmacy, China Medical University, Taichung City 40402, Taiwan;
| | - Zi-Han Gao
- Department of Physiology, National Cheng Kung University Medical College, Tainan City 70101, Taiwan; (Z.-H.G.); (S.-W.L.)
| | - Shih-Wei Li
- Department of Physiology, National Cheng Kung University Medical College, Tainan City 70101, Taiwan; (Z.-H.G.); (S.-W.L.)
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan City 70101, Taiwan; (Z.-H.G.); (S.-W.L.)
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan City 70701, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung City 40402, Taiwan
| |
Collapse
|
11
|
Alcantara-Gonzalez D, Villasana-Salazar B, Peña-Ortega F. Single amyloid-beta injection exacerbates 4-aminopyridine-induced seizures and changes synaptic coupling in the hippocampus. Hippocampus 2019; 29:1150-1164. [PMID: 31381216 DOI: 10.1002/hipo.23129] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/12/2019] [Accepted: 06/05/2019] [Indexed: 11/09/2022]
Abstract
Accumulation of amyloid-beta (Aβ) in temporal lobe structures, including the hippocampus, is related to a variety of Alzheimer's disease symptoms and seems to be involved in the induction of neural network hyperexcitability and even seizures. Still, a direct evaluation of the pro-epileptogenic effects of Aβ in vivo, and of the underlying mechanisms, is missing. Thus, we tested whether the intracisternal injection of Aβ modulates 4-aminopyridine (4AP)-induced epileptiform activity, hippocampal network function, and its synaptic coupling. When tested 3 weeks after its administration, Aβ (but not its vehicle) reduces the latency for 4AP-induced seizures, increases the number of generalized seizures, exacerbates the time to fully recover from seizures, and favors seizure-induced death. These pro-epileptogenic effects of Aβ correlate with a reduction in the power of the spontaneous hippocampal network activity, involving all frequency bands in vivo and only the theta band (4-10 Hz) in vitro. The pro-epileptogenic effects of Aβ also correlate with a reduction of the Schaffer-collateral CA1 synaptic coupling in vitro, which is exacerbated by the sequential bath application of 4-AP and Aβ. In summary, Aβ produces long-lasting pro-epileptic effects that can be due to alterations in the hippocampal circuit, impacting its coordinated network activity and its synaptic efficiency. It is likely that normalizing synaptic coupling and/or coordinated neural network activity (i.e., theta activity) may contribute not only to improve cognitive function in Alzheimer's disease but also to avoid hyperexcitation in conditions of amyloidosis.
Collapse
Affiliation(s)
- David Alcantara-Gonzalez
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Qro, Mexico
| | - Benjamín Villasana-Salazar
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Qro, Mexico
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Qro, Mexico
| |
Collapse
|
12
|
Kreir M, De Bondt A, Van den Wyngaert I, Teuns G, Lu HR, Gallacher DJ. Role of Kv7.2/Kv7.3 and M 1 muscarinic receptors in the regulation of neuronal excitability in hiPSC-derived neurons. Eur J Pharmacol 2019; 858:172474. [PMID: 31238068 DOI: 10.1016/j.ejphar.2019.172474] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 06/17/2019] [Accepted: 06/18/2019] [Indexed: 01/12/2023]
Abstract
The Kv7 family of voltage-dependent non-inactivating potassium channels is composed of five members, of which four are expressed in the CNS. Kv7.2, 7.3 and 7.5 are responsible for the M-current, which plays a critical role in the regulation of neuronal excitability. Stimulation of M1 muscarinic acetylcholine receptor, M1 receptor, increases neuronal excitability by suppressing the M-current generated by the Kv7 channel family. The M-current modulation via M1 receptor is well-described in in vitro assays using cell lines and in native rodent tissue. However, this mechanism was not yet reported in human induced pluripotent stem cells (hiPSC) derived neurons. In the present study, we investigated the effects of both agonists and antagonists of Kv7.2/7.3 channel and M1 receptor in hiPSC derived neurons and in primary rat cortical neuronal cells. The role of M1 receptors in the modulation of neuronal excitability could be demonstrated in both rat primary and hiPSC neurons. The M1 receptors agonist, xanomeline, increased neuronal excitability in both rat cortical and the hiPSC neuronal cells. Furthermore, M1 receptor agonist-induced neuronal excitability in vitro was reduced by an agonist of Kv7.2/7.3 in both neuronal cells. These results show that hiPSC derived neurons recreate the modulation of the M-current by the muscarinic receptor in hiPSC neurons similarly to rat native neurons. Thus, hiPSC neurons could be a useful human-based cell assay for characterization of drugs that affect neuronal excitability and/or induce seizure activity by modulation of M1 receptors or inhibition of Kv7 channels.
Collapse
Affiliation(s)
- Mohamed Kreir
- Non-Clinical Safety, Discovery, Product Development & Supply, Janssen Research and Development, Janssen Pharmaceutica NV, Beerse, Belgium.
| | - An De Bondt
- Computational Sciences, Discovery Sciences, Product Development & Supply, Janssen Research and Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Ilse Van den Wyngaert
- Computational Sciences, Discovery Sciences, Product Development & Supply, Janssen Research and Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Greet Teuns
- Non-Clinical Safety, Discovery, Product Development & Supply, Janssen Research and Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Hua Rong Lu
- Non-Clinical Safety, Discovery, Product Development & Supply, Janssen Research and Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - David J Gallacher
- Non-Clinical Safety, Discovery, Product Development & Supply, Janssen Research and Development, Janssen Pharmaceutica NV, Beerse, Belgium
| |
Collapse
|
13
|
Bradley JA, Luithardt HH, Metea MR, Strock CJ. In Vitro Screening for Seizure Liability Using Microelectrode Array Technology. Toxicol Sci 2019; 163:240-253. [PMID: 29432603 DOI: 10.1093/toxsci/kfy029] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Drug-induced seizure liabilities produce significant compound attrition during drug discovery. Currently available in vitro cytotoxicity assays cannot predict all toxicity mechanisms due to the failure of these assays to predict sublethal target-specific electrophysiological liabilities. Identification of seizurogenic and other electrophysiological effects at early stages of the drug development process is important to ensure that safe candidate compounds can be developed while chemical design is taking place, long before these liabilities are discovered in costly preclinical in vivo studies. The development of a high throughput and reliable in vitro assay to screen compounds for seizure liabilities would de-risk compounds significantly earlier in the drug discovery process and with greater dependability. Here we describe a method for screening compounds that utilizes rat cortical neurons plated onto multiwell microelectrode array plates to identify compounds that cause neurophysiological disruptions. Changes in 12 electrophysiological parameters (spike train descriptors) were measured after application of known seizurogenic compounds and the response pattern was mapped relative to negative controls, vehicle control and neurotoxic controls. Twenty chemicals with a variety of therapeutic indications and targets, including GABAA antagonists, glycine receptor antagonists, ion channel blockers, muscarinic agonist, δ-opioid receptor agonist, dopaminergic D2/adrenergic receptor blocker and nonsteroidal anti-inflammatory drugs, were tested to assess this system. Sixteen of the seventeen seizurogenic/neurotoxic compounds tested positive for seizure liability or neurotoxicity, moreover, different endpoint response patterns for firing rate, burst characteristics and synchrony that distinguished the chemicals into groups relating to target and seizurogenic response emerged from the data. The negative and vehicle control compounds had no effect on neural activity. In conclusion, the multiwell microelectrode array platform using cryopreserved rat cortical neurons is a highly effective high throughput method for reliably screening seizure liabilities within an early de-risking drug development paradigm.
Collapse
Affiliation(s)
| | | | - Monica R Metea
- Cyprotex US, LLC, An Evotec Company, Watertown, Massachusetts
| | | |
Collapse
|
14
|
So EC, Foo NP, Ko SY, Wu SN. Bisoprolol, Known to Be a Selective β₁-Receptor Antagonist, Differentially but Directly Suppresses I K(M) and I K(erg) in Pituitary Cells and Hippocampal Neurons. Int J Mol Sci 2019; 20:657. [PMID: 30717422 PMCID: PMC6386942 DOI: 10.3390/ijms20030657] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 01/27/2019] [Accepted: 01/30/2019] [Indexed: 12/27/2022] Open
Abstract
Bisoprolol (BIS) is a selective antagonist of β₁ adrenergic receptors. We examined the effects of BIS on M-type K⁺ currents (IK(M)) or erg-mediated K⁺ currents (IK(erg)) in pituitary GH3, R1220 cells, and hippocampal mHippoE-14 cells. As GH₃ cells were exposed to BIS, amplitude of IK(M) was suppressed with an IC50 value of 1.21 μM. The BIS-induced suppression of IK(M) amplitude was not affected by addition of isoproterenol or ractopamine, but attenuated by flupirtine or ivabradine. In cell-attached current, BIS decreased the open probability of M-type K⁺ (KM) channels, along with decreased mean opening time of the channel. BIS decreased IK(erg) amplitude with an IC50 value of 6.42 μM. Further addition of PD-118057 attenuated BIS-mediated inhibition of IK(erg). Under current-clamp conditions, BIS depolarization increased the firing of spontaneous action potentials in GH₃ cells; addition of flupirtine, but not ractopamine, reversed BIS-induced firing rate. In R1220 cells, BIS suppressed IK(M); subsequent application of ML-213(Kv7.2 channel activator) reversed BIS-induced suppression of the current. In hippocampal mHippoE-14 neurons, BIS inhibited IK(M) to a greater extent compared to its depressant effect on IK(erg). This demonstrated that in pituitary cells and hippocampal neurons the presence of BIS is capable of directly and differentially suppressing IK(M) and IK(erg), despite its antagonism of β₁-adrenergic receptors.
Collapse
Affiliation(s)
- Edmund Cheung So
- Department of Anesthesia, An Nan Hospital, China Medical University, Tainan 70965, Taiwan.
- Department of Anesthesia, China Medical University, Taichung 40402, Taiwan.
- Graduate Institute of Medical Sciences, Chang Jung Christian University, Tainan 71101, Taiwan.
| | - Ning-Ping Foo
- Department of Anesthesia, An Nan Hospital, China Medical University, Tainan 70965, Taiwan.
- Graduate Institute of Medical Sciences, Chang Jung Christian University, Tainan 71101, Taiwan.
- Department of Emergency Medicine, An Nan Hospital, China Medical University, Tainan 70965, Taiwan.
| | - Shun Yao Ko
- Graduate Institute of Medical Sciences, Chang Jung Christian University, Tainan 71101, Taiwan.
| | - Sheng-Nan Wu
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan 70101, Taiwan.
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan.
| |
Collapse
|
15
|
Fox KCR, Yih J, Raccah O, Pendekanti SL, Limbach LE, Maydan DD, Parvizi J. Changes in subjective experience elicited by direct stimulation of the human orbitofrontal cortex. Neurology 2018; 91:e1519-e1527. [PMID: 30232252 DOI: 10.1212/wnl.0000000000006358] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 07/13/2018] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE We applied direct cortical stimulation (DCS) to the orbitofrontal cortex (OFC) in neurosurgical patients implanted with intracranial electrodes to probe, with high anatomic precision, the causal link between the OFC and human subjective experience. METHODS We administered 272 instances of DCS at 172 OFC sites in 22 patients with intractable focal epilepsy (from 2011 to 2017), none of whom had seizures originating from the OFC. RESULTS Our observations revealed a rich variety of affective, olfactory, gustatory, and somatosensory changes in the subjective domain. Elicited experiences were largely neutral or negatively valenced (e.g., aversive smells and tastes, sadness, and anger). Evidence was found for preferential left lateralization of negatively valenced experiences and strong right lateralization of neutral effects. Moreover, most of the elicited effects were observed after stimulation of OFC tissue around the transverse orbital sulcus, and none were seen in the most anterior aspects of the OFC. CONCLUSIONS Our study yielded 3 central findings: first, a dissociation between the "silent" anterior and nonsilent middle/posterior OFC where stimulation clearly elicits changes in subjective experience; second, evidence that the OFC might play a causal role in integrating affect and multimodal sensory experiences; and third, clear evidence for left lateralization of negatively valenced effects. Our findings provide important information for clinicians treating OFC injury or planning OFC resection and scientists seeking to understand the brain basis for the integration of sensation, cognition, and affect.
Collapse
Affiliation(s)
- Kieran C R Fox
- From the Department of Neurology and Neurological Sciences, Stanford University, CA
| | - Jennifer Yih
- From the Department of Neurology and Neurological Sciences, Stanford University, CA
| | - Omri Raccah
- From the Department of Neurology and Neurological Sciences, Stanford University, CA
| | - Shrita L Pendekanti
- From the Department of Neurology and Neurological Sciences, Stanford University, CA
| | - Lauren E Limbach
- From the Department of Neurology and Neurological Sciences, Stanford University, CA
| | - Daniella D Maydan
- From the Department of Neurology and Neurological Sciences, Stanford University, CA
| | - Josef Parvizi
- From the Department of Neurology and Neurological Sciences, Stanford University, CA.
| |
Collapse
|
16
|
Salgado-Puga K, Rodríguez-Colorado J, Prado-Alcalá RA, Peña-Ortega F. Subclinical Doses of ATP-Sensitive Potassium Channel Modulators Prevent Alterations in Memory and Synaptic Plasticity Induced by Amyloid-β. J Alzheimers Dis 2018; 57:205-226. [PMID: 28222502 DOI: 10.3233/jad-160543] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In addition to coupling cell metabolism and excitability, ATP-sensitive potassium channels (KATP) are involved in neural function and plasticity. Moreover, alterations in KATP activity and expression have been observed in Alzheimer's disease (AD) and during amyloid-β (Aβ)-induced pathology. Thus, we tested whether KATP modulators can influence Aβ-induced deleterious effects on memory, hippocampal network function, and plasticity. We found that treating animals with subclinical doses (those that did not change glycemia) of a KATP blocker (Tolbutamide) or a KATP opener (Diazoxide) differentially restrained Aβ-induced memory deficit, hippocampal network activity inhibition, and long-term synaptic plasticity unbalance (i.e., inhibition of LTP and promotion of LTD). We found that the protective effect of Tolbutamide against Aβ-induced memory deficit was strong and correlated with the reestablishment of synaptic plasticity balance, whereas Diazoxide treatment produced a mild protection against Aβ-induced memory deficit, which was not related to a complete reestablishment of synaptic plasticity balance. Interestingly, treatment with both KATP modulators renders the hippocampus resistant to Aβ-induced inhibition of hippocampal network activity. These findings indicate that KATP are involved in Aβ-induced pathology and they heighten the potential role of KATP modulation as a plausible therapeutic strategy against AD.
Collapse
Affiliation(s)
- Karla Salgado-Puga
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO, México
| | - Javier Rodríguez-Colorado
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO, México
| | - Roberto A Prado-Alcalá
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO, México
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO, México
| |
Collapse
|
17
|
Sampath D, Valdez R, White AM, Raol YH. Anticonvulsant effect of flupirtine in an animal model of neonatal hypoxic-ischemic encephalopathy. Neuropharmacology 2017; 123:126-135. [PMID: 28587899 DOI: 10.1016/j.neuropharm.2017.06.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 05/07/2017] [Accepted: 06/02/2017] [Indexed: 12/20/2022]
Abstract
Research studies suggest that neonatal seizures, which are most commonly associated with hypoxic-ischemic injury, may contribute to brain injury and adverse neurologic outcome. Unfortunately, neonatal seizures are often resistant to treatment with current anticonvulsants. In the present study, we evaluated the efficacy of flupirtine, administered at clinically relevant time-points, for the treatment of neonatal seizures in an animal model of hypoxic-ischemic injury that closely replicates features of the human syndrome. We also compared the efficacy of flupirtine to that of phenobarbital, the current first-line drug for neonatal seizures. Flupirtine is a KCNQ potassium channel opener. KCNQ channels play an important role in controlling brain excitability during early development. In this study, hypoxic-ischemic injury was induced in neonatal rats, and synchronized video-EEG records were acquired at various time-points during the experiment to identify seizures. The results revealed that flupirtine, administered either 5 min after the first electroclinical seizure, or following completion of 2 h of hypoxia, i.e., during the immediate reperfusion period, reduced the number of rats with electroclinical seizures, and also the frequency and total duration of electroclinical seizures. Further, daily dosing of flupirtine decreased the seizure burden over 3 days following HI-induction, and modified the natural evolution of acute seizures. Moreover, compared to a therapeutic dose of phenobarbital, which was modestly effective against electroclinical seizures, flupirtine showed greater efficacy. Our results indicate that flupirtine is an extremely effective treatment for neonatal seizures in rats and provide evidence for a trial of this medication in newborn humans.
Collapse
Affiliation(s)
- Dayalan Sampath
- Department of Pediatrics, Division of Neurology, School of Medicine, Translational Epilepsy Research Program, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Robert Valdez
- Department of Pediatrics, Division of Neurology, School of Medicine, Translational Epilepsy Research Program, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Andrew M White
- Department of Pediatrics, Division of Neurology, School of Medicine, Translational Epilepsy Research Program, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Yogendra H Raol
- Department of Pediatrics, Division of Neurology, School of Medicine, Translational Epilepsy Research Program, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
18
|
Sampath D, Shmueli D, White AM, Raol YH. Flupirtine effectively prevents development of acute neonatal seizures in an animal model of global hypoxia. Neurosci Lett 2015; 607:46-51. [PMID: 26365409 DOI: 10.1016/j.neulet.2015.09.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 08/18/2015] [Accepted: 09/07/2015] [Indexed: 12/16/2022]
Abstract
Current first-line drugs for the treatment of neonatal seizures have limited efficacy and are associated with side effects. Uncontrolled seizures may exacerbate brain injury and contribute to later-life neurological disability. Therefore, it is critical to develop a treatment for neonatal seizures that is effective and safe. In early-life, when the γ-aminobutyric acid (GABA) inhibitory system is not fully developed, potassium channels play an important role in controlling excitability. An earlier study demonstrated that flupirtine, a KCNQ potassium channel opener, is more efficacious than diazepam and phenobarbital for the treatment of chemoconvulsant-induced neonatal seizures. In newborns, seizures are most commonly associated with hypoxic-ischemic encephalopathy (HIE). Thus, in the present study, we examined the efficacy of flupirtine to treat neonatal seizures in an animal model of global hypoxia. Our results showed that flupirtine dose dependently blocks the occurrence of behavioral seizures in pups during hypoxia. Additionally, flupirtine inhibits the development of hypoxia-induced clinical seizures and associated epileptiform discharges, as well as purely electrographic (subclinical) seizures. These results suggest that flupirtine is an effective anti-seizure drug, and that further studies should be conducted to determine the time window within which it's administration can effectively treat neonatal seizures.
Collapse
Affiliation(s)
- Dayalan Sampath
- Department of Pediatrics, Division of Neurology, School of Medicine, Translational Epilepsy Research Program, University of Colorado, School of Medicine, Aurora, CO 80045, United States
| | - Doron Shmueli
- Department of Pediatrics, Division of Neurology, School of Medicine, Translational Epilepsy Research Program, University of Colorado, School of Medicine, Aurora, CO 80045, United States
| | - Andrew M White
- Department of Pediatrics, Division of Neurology, School of Medicine, Translational Epilepsy Research Program, University of Colorado, School of Medicine, Aurora, CO 80045, United States
| | - Yogendra H Raol
- Department of Pediatrics, Division of Neurology, School of Medicine, Translational Epilepsy Research Program, University of Colorado, School of Medicine, Aurora, CO 80045, United States.
| |
Collapse
|
19
|
Zavala-Tecuapetla C, Tapia D, Rivera-Angulo AJ, Galarraga E, Peña-Ortega F. Morphological characterization of respiratory neurons in the pre-Bötzinger complex. PROGRESS IN BRAIN RESEARCH 2014; 209:39-56. [PMID: 24746042 DOI: 10.1016/b978-0-444-63274-6.00003-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Although the pre-Bötzinger complex (preBötC) was defined as the inspiratory rhythm generator long ago, the functional-anatomical characterization of its neuronal components is still being achieved. Recent advances have identified the expression of molecular markers in the preBötC neurons that, however, are not exclusive to specific respiratory neuron subtypes and have not always been related to specific cell morphologies. Here, we evaluated the morphology and the axonal projections of electrophysiologically defined respiratory neurons in the preBötC using whole-cell recordings and intracellular biocytin labeling. We found that respiratory pacemaker neurons are larger than expiratory neurons and that inspiratory neurons are smaller than pacemaker and expiratory neurons. Other morphological features such as somata shapes or dendritic branching patterns were not found to be significantly different among the preBötC neurons sampled. We also found that both pacemaker and inspiratory nonpacemaker neurons, but not expiratory neurons, show extensive axonal projections to the contralateral preBötC and show signs of electrical coupling. Overall, our data suggest that there are morphological differences between subtypes of preBötC respiratory neurons. It will be important to take such differences in consideration since morphological differences would influence synaptic responses and action potential propagation.
Collapse
Affiliation(s)
- Cecilia Zavala-Tecuapetla
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, UNAM Campus Juriquilla, Querétaro, Mexico; Laboratorio de Nanotecnología, Instituto Nacional de Neurología y Neurocirugía-MVS, Mexico D.F., Mexico; Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados Sede Sur, Mexico D.F., Mexico
| | - Dagoberto Tapia
- Departamento de Biofísica, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico D.F., Mexico
| | - Ana Julia Rivera-Angulo
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, UNAM Campus Juriquilla, Querétaro, Mexico
| | - Elvira Galarraga
- Departamento de Biofísica, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico D.F., Mexico
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, UNAM Campus Juriquilla, Querétaro, Mexico.
| |
Collapse
|
20
|
Rivera-Angulo AJ, Peña-Ortega F. Isocitrate supplementation promotes breathing generation, gasping, and autoresuscitation in neonatal mice. J Neurosci Res 2013; 92:375-88. [DOI: 10.1002/jnr.23330] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 10/15/2013] [Accepted: 10/25/2013] [Indexed: 02/01/2023]
Affiliation(s)
- Ana-Julia Rivera-Angulo
- Departamento de Neurobiología del Desarrollo y Neurofisiología; Instituto de Neurobiología; Universidad Nacional Autónoma de México-Campus Juriquilla; Querétaro México
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología; Instituto de Neurobiología; Universidad Nacional Autónoma de México-Campus Juriquilla; Querétaro México
| |
Collapse
|
21
|
Shah NH, Aizenman E. Voltage-gated potassium channels at the crossroads of neuronal function, ischemic tolerance, and neurodegeneration. Transl Stroke Res 2013; 5:38-58. [PMID: 24323720 DOI: 10.1007/s12975-013-0297-7] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 09/14/2013] [Accepted: 10/14/2013] [Indexed: 11/29/2022]
Abstract
Voltage-gated potassium (Kv) channels are widely expressed in the central and peripheral nervous system and are crucial mediators of neuronal excitability. Importantly, these channels also actively participate in cellular and molecular signaling pathways that regulate the life and death of neurons. Injury-mediated increased K(+) efflux through Kv2.1 channels promotes neuronal apoptosis, contributing to widespread neuronal loss in neurodegenerative disorders such as Alzheimer's disease and stroke. In contrast, some forms of neuronal activity can dramatically alter Kv2.1 channel phosphorylation levels and influence their localization. These changes are normally accompanied by modifications in channel voltage dependence, which may be neuroprotective within the context of ischemic injury. Kv1 and Kv7 channel dysfunction leads to neuronal hyperexcitability that critically contributes to the pathophysiology of human clinical disorders such as episodic ataxia and epilepsy. This review summarizes the neurotoxic, neuroprotective, and neuroregulatory roles of Kv channels and highlights the consequences of Kv channel dysfunction on neuronal physiology. The studies described in this review thus underscore the importance of normal Kv channel function in neurons and emphasize the therapeutic potential of targeting Kv channels in the treatment of a wide range of neurological diseases.
Collapse
Affiliation(s)
- Niyathi Hegde Shah
- Department of Neurobiology, University of Pittsburgh School of Medicine, 3500 Terrace Street, E1456 BST, Pittsburgh, PA, 15261, USA,
| | | |
Collapse
|
22
|
Maslarova A, Salar S, Lapilover E, Friedman A, Veh RW, Heinemann U. Increased susceptibility to acetylcholine in the entorhinal cortex of pilocarpine-treated rats involves alterations in KCNQ channels. Neurobiol Dis 2013; 56:14-24. [DOI: 10.1016/j.nbd.2013.02.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 02/19/2013] [Accepted: 02/25/2013] [Indexed: 02/03/2023] Open
|
23
|
Lasoń W, Chlebicka M, Rejdak K. Research advances in basic mechanisms of seizures and antiepileptic drug action. Pharmacol Rep 2013; 65:787-801. [DOI: 10.1016/s1734-1140(13)71060-0] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 03/11/2013] [Indexed: 10/25/2022]
|
24
|
Amyloid Beta peptides differentially affect hippocampal theta rhythms in vitro. INTERNATIONAL JOURNAL OF PEPTIDES 2013; 2013:328140. [PMID: 23878547 PMCID: PMC3708430 DOI: 10.1155/2013/328140] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Accepted: 06/03/2013] [Indexed: 12/27/2022]
Abstract
Soluble amyloid beta peptide (A β ) is responsible for the early cognitive dysfunction observed in Alzheimer's disease. Both cholinergically and glutamatergically induced hippocampal theta rhythms are related to learning and memory, spatial navigation, and spatial memory. However, these two types of theta rhythms are not identical; they are associated with different behaviors and can be differentially modulated by diverse experimental conditions. Therefore, in this study, we aimed to investigate whether or not application of soluble A β alters the two types of theta frequency oscillatory network activity generated in rat hippocampal slices by application of the cholinergic and glutamatergic agonists carbachol or DHPG, respectively. Due to previous evidence that oscillatory activity can be differentially affected by different A β peptides, we also compared Aβ 25-35 and Aβ 1-42 for their effects on theta rhythms in vitro at similar concentrations (0.5 to 1.0 μ M). We found that Aβ 25-35 reduces, with less potency than Aβ 1-42, carbachol-induced population theta oscillatory activity. In contrast, DHPG-induced oscillatory activity was not affected by a high concentration of Aβ 25-35 but was reduced by Aβ 1-42. Our results support the idea that different amyloid peptides might alter specific cellular mechanisms related to the generation of specific neuronal network activities, instead of exerting a generalized inhibitory effect on neuronal network function.
Collapse
|
25
|
Amyloid Beta-Protein and Neural Network Dysfunction. JOURNAL OF NEURODEGENERATIVE DISEASES 2013; 2013:657470. [PMID: 26316994 PMCID: PMC4437331 DOI: 10.1155/2013/657470] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2012] [Accepted: 12/06/2012] [Indexed: 01/15/2023]
Abstract
Understanding the neural mechanisms underlying brain dysfunction induced by amyloid beta-protein (Aβ) represents one of the major challenges for Alzheimer's disease (AD) research. The most evident symptom of AD is a severe decline in cognition. Cognitive processes, as any other brain function, arise from the activity of specific cell assemblies of interconnected neurons that generate neural network dynamics based on their intrinsic and synaptic properties. Thus, the origin of Aβ-induced cognitive dysfunction, and possibly AD-related cognitive decline, must be found in specific alterations in properties of these cells and their consequences in neural network dynamics. The well-known relationship between AD and alterations in the activity of several neural networks is reflected in the slowing of the electroencephalographic (EEG) activity. Some features of the EEG slowing observed in AD, such as the diminished generation of different network oscillations, can be induced in vivo and in vitro upon Aβ application or by Aβ overproduction in transgenic models. This experimental approach offers the possibility to study the mechanisms involved in cognitive dysfunction produced by Aβ. This type of research may yield not only basic knowledge of neural network dysfunction associated with AD, but also novel options to treat this modern epidemic.
Collapse
|
26
|
Chege SW, Hortopan GA, T Dinday M, Baraban SC. Expression and function of KCNQ channels in larval zebrafish. Dev Neurobiol 2012; 72:186-98. [PMID: 21692188 DOI: 10.1002/dneu.20937] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Members of the K(v)7 family generate a subthreshold potassium current, termed M-current, that regulates the excitability of principal central neurons. Mutations in two members of this family, K(v)7.2 (KCNQ2) and K(v)7.3 (KCNQ3) are associated with a neurological disorder known as benign familial neonatal convulsion (BFNC). Despite their importance in normal and pathological brain function, developmental expression and function of these channels remains relatively unexplored. Here, we examined the temporal expression of K(v)7 channel subunits in zebrafish larvae using a real-time quantitative PCR approach. Spatial expression in the larval zebrafish brain was assessed using whole-mount in situ hybridization. The mRNA for three members of the K(v)7 family (KCNQ2, 3 and 5) is reported in zebrafish between two and seven days post-fertilization (dpf). Using electrophysiological techniques, we show that inhibitors of K(v)7 channels (linopirdine and XE991) induce burst discharge activity in immature zebrafish between 3 and 7 dpf. This abnormal electrical activity is blocked by a K(v)7 channel opener (retigabine) and was also shown to evoke convulsive behaviors in freely swimming zebrafish. Using morpholino oligonucleotides directed against KCNQ3, we confirmed a role for KCNQ channels in generation of electrical burst discharges. These results indicate that functional K(v)7 channels are expressed in the larval zebrafish nervous system and could play a direct role in generation of seizure activity.
Collapse
Affiliation(s)
- Sally W Chege
- PIBS Graduate Program in Neuroscience, University of California, San Francisco, San Francisco, California 94143, USA
| | | | | | | |
Collapse
|
27
|
Molinari F, Cattani A, Mdzomba J, Aniksztejn L. Glutamate transporters control metabotropic glutamate receptors activation to prevent the genesis of paroxysmal burst in the developing hippocampus. Neuroscience 2012; 207:25-36. [DOI: 10.1016/j.neuroscience.2012.01.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Revised: 12/23/2011] [Accepted: 01/19/2012] [Indexed: 12/26/2022]
|
28
|
Ramírez-Jarquín JO, Lara-Hernández S, López-Guerrero JJ, Aguileta MA, Rivera-Angulo AJ, Sampieri A, Vaca L, Ordaz B, Peña-Ortega F. Somatostatin modulates generation of inspiratory rhythms and determines asphyxia survival. Peptides 2012; 34:360-72. [PMID: 22386651 DOI: 10.1016/j.peptides.2012.02.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 02/15/2012] [Accepted: 02/15/2012] [Indexed: 10/28/2022]
Abstract
Breathing and the activity of its generator (the pre-Bötzinger complex; pre-BötC) are highly regulated functions. Among neuromodulators of breathing, somatostatin (SST) is unique: it is synthesized by a subset of glutamatergic pre-BötC neurons, but acts as an inhibitory neuromodulator. Moreover, SST regulates breathing both in normoxic and in hypoxic conditions. Although it has been implicated in the neuromodulation of breathing, neither the locus of SST modulation, nor the receptor subtypes involved have been identified. In this study, we aimed to fill in these blanks by characterizing the SST-induced regulation of inspiratory rhythm generation in vitro and in vivo. We found that both endogenous and exogenous SST depress all preBötC-generated rhythms. While SST abolishes sighs, it also decreases the frequency and increases the regularity of eupnea and gasping. Pharmacological experiments showed that SST modulates inspiratory rhythm generation by activating SST receptor type-2, whose mRNA is abundantly expressed in the pre-Bötzinger complex. In vivo, blockade of SST receptor type-2 reduces gasping amplitude and consequently, it precludes auto-resuscitation after asphyxia. Based on our findings, we suggest that SST functions as an inhibitory neuromodulator released by excitatory respiratory neurons when they become overactivated in order to stabilize breathing rhythmicity in normoxic and hypoxic conditions.
Collapse
Affiliation(s)
- Josué O Ramírez-Jarquín
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, UNAM-Campus Juriquilla, Mexico.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Experimental models of seizures and epilepsies. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 105:57-82. [PMID: 22137429 DOI: 10.1016/b978-0-12-394596-9.00003-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Epilepsy is one of the most common neurological conditions that affect people of all ages. Epilepsy is characterized by occurrence of spontaneous recurrent seizures. Currently available drugs are ineffective in controlling seizures in approximately one-third of patients with epilepsy. Moreover, these drugs are associated with adverse effects, and none of them are effective in preventing development of epilepsy following an insult or injury. To develop an effective therapeutic strategy that can interfere with the process of development of epilepsy (epileptogenesis), it is crucial to study the changes that occur in the brain after an injury and before epilepsy develops. It is not possible to determine these changes in human tissue for obvious ethical reasons. Over the years, experimental models of epilepsies have contributed immensely in improving our understanding of mechanism of epileptogenesis as well as of seizure generation. There are many models that replicate at least some of the characteristics of human epilepsy. Each model has its advantages and disadvantages, and the investigator should be aware of this before selecting a specific model for his/her studies. Availability of a good animal model is a key to the development of an effective treatment. Unfortunately, there are many epilepsy syndromes, specifically pediatric, which still lack a valid animal model. It is vital that more research is done to develop animal models for such syndromes.
Collapse
|
30
|
Guan D, Higgs MH, Horton LR, Spain WJ, Foehring RC. Contributions of Kv7-mediated potassium current to sub- and suprathreshold responses of rat layer II/III neocortical pyramidal neurons. J Neurophysiol 2011; 106:1722-33. [PMID: 21697446 DOI: 10.1152/jn.00211.2011] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
After block of Kv1- and Kv2-mediated K(+) currents in acutely dissociated neocortical pyramidal neurons from layers II/III of rat somatosensory and motor cortex, the remaining current is slowly activating and persistent. We used whole cell voltage clamp to show that the Kv7 blockers linopirdine and XE-991 blocked a current with similar kinetics to the current remaining after combined block of Kv1 and Kv2 channels. This current was sensitive to low doses of linopirdine and activated more slowly and at more negative potentials than Kv1- or Kv2-mediated current. The Kv7-mediated current decreased in amplitude with time in whole cell recordings, but in most cells the current was stable for several minutes. Current in response to a traditional M-current protocol was blocked by muscarine, linopirdine, and XE-991. Whole cell slice recordings revealed that the Q₁₀ for channel deactivation was ∼2.5. Sharp electrode current-clamp recordings from adult pyramidal cells demonstrated that block of Kv7-mediated current with XE-991 reduced rheobase, shortened the latency to firing to near rheobase current, induced more regular firing at low current intensity, and increased the rate of firing to a given current injection. XE-991 did not affect single action potentials or spike frequency adaptation. Application of XE-991 also eliminated subthreshold voltage oscillations and increased gain for low-frequency inputs (<10 Hz) without affecting gain for higher frequency inputs. These data suggest important roles for Kv7 channels in subthreshold regulation of excitability, generation of theta-frequency subthreshold oscillations, regulation of interspike intervals, and biasing selectivity toward higher frequency inputs.
Collapse
Affiliation(s)
- D Guan
- Department of Anatomy and Neurobiology, University of Tennessee, Memphis, TN 38163, USA
| | | | | | | | | |
Collapse
|
31
|
Barrese V, Miceli F, Soldovieri MV, Ambrosino P, Iannotti FA, Cilio MR, Taglialatela M. Neuronal potassium channel openers in the management of epilepsy: role and potential of retigabine. Clin Pharmacol 2010; 2:225-36. [PMID: 22291509 PMCID: PMC3262367 DOI: 10.2147/cpaa.s15369] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Despite the availability of over 20 antiepileptic drugs, about 30% of epileptic patients do not achieve seizure control. Thus, identification of additional molecules targeting novel molecular mechanisms is a primary effort in today's antiepileptic drug research. This paper reviews the pharmacological development of retigabine, an antiepileptic drug with a novel mechanism of action, namely the activation of voltage-gated potassium channels of the Kv7 subfamily. These channels, which act as widespread regulators of intrinsic neuronal excitability and of neurotransmitter-induced network excitability changes, are currently viewed among the most promising targets for anticonvulsant pharmacotherapy. In particular, the present work reviews the pathophysiological role of Kv7 channels in neuronal function, the molecular mechanisms involved in the Kv7 channel-opening action of retigabine, the activity of retigabine in preclinical in vitro and in vivo studies predictive of anticonvulsant activities, and the clinical status of development for this drug as an add-on treatment for pharmacoresistant epilepsy. Particular efforts are devoted to highlighting the potential advantages and disadvantages of retigabine when compared with currently available compounds, in order to provide a comprehensive assessment of its role in therapy for treatment-resistant epilepsies.
Collapse
Affiliation(s)
- Vincenzo Barrese
- Department of Neuroscience, University of Naples Federico II, Naples
| | | | | | | | | | | | | |
Collapse
|
32
|
Adaya-Villanueva A, Ordaz B, Balleza-Tapia H, Márquez-Ramos A, Peña-Ortega F. Beta-like hippocampal network activity is differentially affected by amyloid beta peptides. Peptides 2010; 31:1761-6. [PMID: 20558221 DOI: 10.1016/j.peptides.2010.06.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2009] [Revised: 06/05/2010] [Accepted: 06/06/2010] [Indexed: 01/29/2023]
Abstract
Alzheimer disease (AD) patients show alterations in both neuronal network oscillations and the cognitive processes associated to them. Related to this clinical observation, it has been found that amyloid beta protein (Abeta) differentially affects some hippocampal network activities, reducing theta and gamma oscillations, without affecting sharp waves and ripples. Beta-like oscillations is another cognitive-related network activity that can be evoked in hippocampal slices by several experimental manipulations, including bath application of kainate and increasing extracellular potassium. Here, we tested whether or not different Abeta peptides differentially affect beta-like oscillatory patterns. We specifically tested the effects of fresh dissolved Abeta(25-35) and oligomerized Abeta(1-42) and found that kainate-induced oscillatory network activity was affected, in a slightly concentration dependent-manner, by both fresh dissolved (mostly monomeric) Abeta(25-35) and oligomeric Abeta(1-42). In contrast, potassium-induced oscillatory activity, which is reduced by oligomeric Abeta(1-42), is not affected by monomeric Abeta(25-35) at any of the concentrations tested. Our results support the idea that different amyloid peptides might alter specific cellular mechanisms related to the generation of specific neuronal network activities, instead of a generalized inhibitory effect of Abeta peptides on neuronal network function.
Collapse
|
33
|
Peña F, Ordaz B, Balleza-Tapia H, Bernal-Pedraza R, Márquez-Ramos A, Carmona-Aparicio L, Giordano M. Beta-amyloid protein (25-35) disrupts hippocampal network activity: role of Fyn-kinase. Hippocampus 2010; 20:78-96. [PMID: 19294646 DOI: 10.1002/hipo.20592] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Early cognitive deficit characteristic of early Alzheimer's disease seems to be produced by the soluble forms of beta-amyloid protein. Such cognitive deficit correlates with neuronal network dysfunction that is reflected as alterations in the electroencephalogram of both Alzheimer patients and transgenic murine models of such disease. Correspondingly, recent studies have demonstrated that chronic exposure to betaAP affects hippocampal oscillatory properties. However, it is still unclear if such neuronal network dysfunction results from a direct action of betaAP on the hippocampal circuit or it is secondary to the chronic presence of the protein in the brain. Therefore, we aimed to explore the effect of acute exposure to betaAP(25-35) on hippocampal network activity both in vitro and in vivo, as well as on intrinsic and synaptic properties of hippocampal neurons. We found that betaAP(25-35), reversibly, affects spontaneous hippocampal population activity in vitro. Such effect is not produced by the inverse sequence betaAP(35-25) and is reproduced by the full-length peptide betaAP(1-42). Correspondingly betaAP(25-35), but not the inverse sequence betaAP(35-25), reduces theta-like activity recorded from the hippocampus in vivo. The betaAP(25-35)-induced disruption in hippocampal network activity correlates with a reduction in spontaneous neuronal activity and synaptic transmission, as well as with an inhibition in the subthreshold oscillations produced by pyramidal neurons in vitro. Finally, we studied the involvement of Fyn-kinase on the betaAP(25-35)-induced disruption in hippocampal network activity in vitro. Interestingly, we found that such phenomenon is not observed in slices obtained from Fyn-knockout mice. In conclusion, our data suggest that betaAP acutely affects proper hippocampal function through a Fyn-dependent mechanism. We propose that such alteration might be related to the cognitive impairment observed, at least, during the early phases of Alzheimer's disease.
Collapse
Affiliation(s)
- Fernando Peña
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados Sede Sur, México, D.F., México.
| | | | | | | | | | | | | |
Collapse
|
34
|
Peña F. PACAP modulates the respiratory rhythm generated in the brainstem slice preparation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 669:119-22. [PMID: 20217333 DOI: 10.1007/978-1-4419-5692-7_24] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is involved in breathing control and its absence leads to sudden death of neonatal mice. It has also been shown that exogenous PACAP application increases breathing by activating peripheral chemoreceptors. However, the contribution of the central respiratory network to PACAP activation of breathing has not been yet established. Here, I explore the effect of PACAP on the rhythmic inspiratory activity generated by the preBötzinger complex (PreBötC) in the brainstem slice preparation. The results show that PACAP increases, in a concentration-dependent manner, burst frequency and decreases burst amplitude of the respiratory rhythm generated in vitro. In conclusion, PACAP is a neuromodulator of the PreBötC suggesting that alterations in such neuromodulation might be involved in the sudden infant death-like phenotype.
Collapse
Affiliation(s)
- Fernando Peña
- Departamento de Farmacobiología, Cinvestav-IPN, México, DF, México.
| |
Collapse
|
35
|
Raol YH, Lapides DA, Keating JG, Brooks-Kayal AR, Cooper EC. A KCNQ channel opener for experimental neonatal seizures and status epilepticus. Ann Neurol 2009; 65:326-36. [PMID: 19334075 DOI: 10.1002/ana.21593] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Neonatal seizures occur frequently, are often refractory to anticonvulsants, and are associated with considerable morbidity and mortality. Genetic and electrophysiological evidence indicates that KCNQ voltage-gated potassium channels are critical regulators of neonatal brain excitability. This study tests the hypothesis that selective openers of KCNQ channels may be effective for treatment of neonatal seizures. METHODS We induced seizures in postnatal day 10 rats with either kainic acid or flurothyl. We measured seizure activity using quantified behavioral rating and electrocorticography. We compared the efficacy of flupirtine, a selective KCNQ channel opener, with phenobarbital and diazepam, two drugs in current use for neonatal seizures. RESULTS Unlike phenobarbital or diazepam, flupirtine prevented animals from experiencing development of status epilepticus when administered before kainate. In the flurothyl model, phenobarbital and diazepam increased latency to seizure onset, but flupirtine completely prevented seizures throughout the experiment. Flupirtine was also effective in arresting electrographic and behavioral seizures when administered after animals had developed continuous kainate-induced status epilepticus. Flupirtine caused dose-related sedation and suppressed electroencephalographic activity but did not result in respiratory suppression or result in any mortality. INTERPRETATION Flupirtine appears more effective than either of two commonly used antiepileptic drugs, phenobarbital and diazepam, in preventing and suppressing seizures in both the kainic acid and flurothyl models of symptomatic neonatal seizures. KCNQ channel openers merit further study as potential treatments for seizures in infants and children.
Collapse
Affiliation(s)
- Yogendrasinh H Raol
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
36
|
Mazarati A, Wu J, Shin D, Kwon YS, Sankar R. Antiepileptogenic and antiictogenic effects of retigabine under conditions of rapid kindling: an ontogenic study. Epilepsia 2008; 49:1777-86. [PMID: 18503560 DOI: 10.1111/j.1528-1167.2008.01674.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE To examine antiepileptogenic and antiictogenic potential of retigabine (RTG) under conditions of rapid kindling epileptogenesis during different stages of development. METHODS The experiments were performed in postnatal day 14 (P14), P21, and P35 male Wistar rats. After stereotaxic implantation of hippocampal stimulating and recording electrodes, the effects of RTG on baseline afterdischarge (AD) properties were studied. Next, the animals underwent rapid kindling (sixty 10 s trains, bipolar 20 Hz square wave pulses delivered every 5 min). The progression of seizures (kindling acquisition), and responses to test stimulations after kindling (retention) were compared between RTG and vehicle-treated rats. Additionally, the effects of RTG on the severity of seizures in previously kindled animals were examined. RESULTS When administered intraperitoneally in doses that induced only mild, or no motor deficits, RTG significantly dampened brain excitability, evident as the increase of AD threshold and shortening of AD duration. During kindling, RTG delayed the development of focal seizures in P14 rats, and prevented the occurrence of full limbic seizures at all three ages. At P14 and P21, but not at P35, pretreatment with RTG prevented the establishment of kindling-induced enhanced seizure susceptibility. Administration of RTG to kindled animals decreased the severity of seizures induced by test stimulation. The effect was most prominent at P14. DISCUSSION RTG exerted both antiepileptogenic and antiictogenic effects under conditions of rapid kindling model. These effects were apparent during postneonatal, early childhood, and adolescent stages of development.
Collapse
Affiliation(s)
- Andréy Mazarati
- Department of Pediatrics, Neurology Division, David Geffen School of Medicine at UCLA, Los Angeles, California 90095-1752, USA.
| | | | | | | | | |
Collapse
|
37
|
Zavala-Tecuapetla C, Aguileta MA, Lopez-Guerrero JJ, González-Marín MC, Peña F. Calcium-activated potassium currents differentially modulate respiratory rhythm generation. Eur J Neurosci 2008; 27:2871-84. [PMID: 18445052 DOI: 10.1111/j.1460-9568.2008.06214.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The pre-Bötzinger complex (PBC) generates eupnea and sighs in normoxia and gasping during hypoxia through particular mixtures of intrinsic and synaptic properties. Among intrinsic properties, little is known about the role of Ca(2+)-activated potassium channels in respiratory rhythms generation. To examine this role, we tested the effects of openers and blockers of the large-conductance (BK) and small-conductance (SK) Ca(2+)-activated potassium channels on the respiratory rhythms recorded both in vitro and in vivo, as well as on the discharge pattern of respiratory neurons in the PBC. Activation of SK channels with 1-ethyl-2-benzimidazolinone (1-EBIO) abolished sigh-like activity and inhibited eupneic-like activity, whereas blockade of SK channels with apamine (APA) increased frequency in both rhythms. In hypoxia, APA did not affect the transition to gasping-like activity. At the cellular level, activation of SK channels abolished pacemaker activity and decreased non-pacemaker neurons discharge; opposite effects were observed with SK blockade. In contrast to SK channel modulation, either activation or blockade of BK channels with NS 1619 or iberiotoxin and paxilline, respectively, produced mild effects on eupneic-like and sigh-like bursts during normoxia in vitro. However, BK blockers prevented the changes associated with the transition to gasping-like activity in vitro and perturbed gasping generation and autoresuscitation in vivo. At the cellular level BK channel modulation did not affect respiratory neurons discharge. We conclude that K(Ca) participate in rhythm generation in a state-dependent manner; SK channels are preferentially involved in rhythm generation in normoxia whereas BK channels participate in the transition to gasping generation in hypoxia.
Collapse
Affiliation(s)
- C Zavala-Tecuapetla
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del I.P.N., México, DF, México
| | | | | | | | | |
Collapse
|
38
|
Standard antiepileptic drugs fail to block epileptiform activity in rat organotypic hippocampal slice cultures. Br J Pharmacol 2008; 154:709-24. [PMID: 18414393 DOI: 10.1038/bjp.2008.112] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND AND PURPOSE Earlier studies had demonstrated that tonic-clonic seizure-like events (SLEs) resembling electrographic correlates of limbic seizures in animals and humans can be induced in organotypic hippocampal slice cultures (OHSCs). We have explored OHSCs for their suitability to serve as in vitro models of limbic seizures for studying seizure mechanisms and screening new antiepileptic compounds. EXPERIMENTAL APPROACH OHSCs were cultivated according to the interface method. Neuronal activity and extracellular potassium concentration were recorded under submerged conditions. SLEs were induced by lowering magnesium concentration or by applying the potassium channel blocker 4-aminopyridine. The effects of standard antiepileptic drugs (AEDs), carbamazepine, phenytoin, valproic acid, clonazepam, diazepam and phenobarbital sodium on SLEs were analysed. KEY RESULTS In more than 93% of OHSCs, AEDs did not prevent the induction of SLEs or stop ongoing seizure activity even when toxic concentrations were applied. This pharmacoresistance was independent of the method of seizure provocation, postnatal age at explantation (P2-P10) and cultivation time in vitro (2 months). SLEs were reversibly blocked by glutamate antagonists or the GABA(A)-agonist muscimol. CONCLUSIONS AND IMPLICATIONS We present a simple to establish in vitro model of tonic-clonic SLEs that is a priori pharmacoresistant and thus has an advantage over animal models of pharmacoresistant seizures in which responders and non-responders can be sorted out only after an experiment. OHSCs could be suitable for exploring mechanisms of pharmacoresistant seizures and be used for the identification of new anticonvulsive compounds eventually effective in drug refractory epilepsy.
Collapse
|
39
|
Young SR, Bianchi R, Wong RKS. Signaling mechanisms underlying group I mGluR-induced persistent AHP suppression in CA3 hippocampal neurons. J Neurophysiol 2008; 99:1105-18. [PMID: 18184892 DOI: 10.1152/jn.00435.2007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Activation of group I metabotropic glutamate receptors (mGluRs) leads to a concerted modulation of spike afterpotentials in guinea pig hippocampal neurons including a suppression of both medium and slow afterhyperpolarizations (AHPs). Suppression of AHPs may be long-lasting, in that it persists after washout of the agonist. Here, we show that persistent AHP suppression differs from short-term, transient suppression in that distinct and additional signaling processes are required to render the suppression persistent. Persistent AHP suppression followed DHPG application for 30 min, but not DHPG application for 5 min. Persistent AHP suppression was temperature dependent, occurring at 30-31 degrees C, but not at 25-26 degrees C. Preincubation of slices in inhibitors of protein synthesis (cycloheximide or anisomycin) prevented the persistent suppression of AHPs by DHPG. Similarly, preincubation of slices in an inhibitor of p38 MAP kinase (SB 203580) prevented persistent AHP suppression. In contrast, a blocker of p42/44 MAP kinase activation (PD 98059) had no effect on persistent AHP suppression. Additionally, we show that the mGluR5 antagonist MPEP, but not the mGluR1 antagonist LY 367385, prevented DHPG-induced persistent AHP suppression. Thus persistent AHP suppression by DHPG in hippocampal neurons requires activation of mGluR5. In addition, activation of p38 MAP kinase signaling and protein synthesis are required to impart persistence to the DHPG-activated AHP suppression.
Collapse
Affiliation(s)
- Steven R Young
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY 11203, USA.
| | | | | |
Collapse
|
40
|
Qiu C, Johnson BN, Tallent MK. K+ M-current regulates the transition to seizures in immature and adult hippocampus. Epilepsia 2007; 48:2047-58. [PMID: 17651418 DOI: 10.1111/j.1528-1167.2007.01193.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
PURPOSE Loss-of-function mutations in Kv7.2 or Kv7.3 K(+) channel subunits underlies the neonatal epilepsy benign familial neonatal convulsions (BFNC). These two subunits interact to form a functional K(+) channel that underlies the M-current (I(M)), a voltage-dependent noninactivating K(+) current. In BFNC, seizures begin shortly after birth, and spontaneously remit in the first few months of life. The nature of this window of vulnerability is unclear. We address this issue using a hippocampal slice model, to study the effects of I(M) blockade or augmentation on epileptiform activity. METHODS We used the Mg(+)(+)-free seizure model in adult and immature (P8-P15) acute rat hippocampal slices. We recorded from both CA1 and CA3 regions using extracellular and intracellular methods. RESULTS When M-channels are blocked pharmacologically, the transition from interictal to ictal bursting becomes much more likely, especially in immature brain. We also show augmentation of I(M) is effective in stopping ictal events in immature brain, at the developmental age that approximates a human newborn in cortical development. I(M) appears to counter the sustained N-methyl-D-aspartate (NMDA) receptor-mediated depolarizations needed to trigger an ictal event. The increased likelihood of ictal bursting by I(M) blockade is not shared by other selective K(+) channel blockers that increase hippocampal excitability. CONCLUSIONS Voltage-dependent M-channels are activated during interictal bursts and contribute to burst termination. When these channels are compromised, interictal burst duration becomes sufficient to trigger the sustained depolarizations that underlie ictal bursts. This transition to ictal bursts upon I(M) blockade is especially likely to occur in immature hippocampus. This selective function of M-channels likely contributes to the transient window of vulnerability to seizures that occurs with BFNC.
Collapse
Affiliation(s)
- Cuie Qiu
- Drexel University College of Medicine, 245 15th Street, Philadelphia, PA 19348, U.S.A
| | | | | |
Collapse
|
41
|
Kalueff AV, Wheaton M, Murphy DL. What's wrong with my mouse model? Behav Brain Res 2007; 179:1-18. [PMID: 17306892 DOI: 10.1016/j.bbr.2007.01.023] [Citation(s) in RCA: 208] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2006] [Revised: 01/15/2007] [Accepted: 01/19/2007] [Indexed: 10/23/2022]
Abstract
Stress plays a key role in pathogenesis of anxiety and depression. Animal models of these disorders are widely used in behavioral neuroscience to explore stress-evoked brain abnormalities, screen anxiolytic/antidepressant drugs and establish behavioral phenotypes of gene-targeted or transgenic animals. Here we discuss the current situation with these experimental models, and critically evaluate the state of the art in this field. Noting a deficit of fresh ideas and especially new paradigms for animal anxiety and depression models, we review existing challenges and outline important directions for further research in this field.
Collapse
Affiliation(s)
- A V Kalueff
- Laboratory of Clinical Science, National Institute of Mental Health, Bethesda, MD 20892-1264, USA.
| | | | | |
Collapse
|
42
|
Peña F, Aguileta MA. Effects of riluzole and flufenamic acid on eupnea and gasping of neonatal mice in vivo. Neurosci Lett 2007; 415:288-93. [PMID: 17276002 DOI: 10.1016/j.neulet.2007.01.032] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2006] [Revised: 01/13/2007] [Accepted: 01/17/2007] [Indexed: 10/23/2022]
Abstract
The pre-Bötzinger complex (PBC), part of the ventral respiratory group that is responsible for inspiratory rhythm generation, contains at least two types of pacemaker neurons. In vitro studies have shown that bursting properties of one type of pacemaker relies on a riluzole-sensitive persistent sodium current, whereas bursting of a second type is sensitive to flufenamic acid (FFA), a calcium-dependent nonspecific cationic current blocker. In vitro, under control conditions, the PBC generates fictive eupneic activity that depends on both riluzole-sensitive and FFA-sensitive pacemaker neurons. During hypoxia the PBC generates fictive gasping activity and only riluzole-sensitive pacemaker neurons appear to be necessary for this rhythm. We carried out pharmacological experiments to test the role of respiratory pacemaker neurons in vivo by performing plethysmographic recordings on neonate mice. As reported in vitro, eupnea activity in vivo is abolished only if both FFA and riluzole are coadministered intracisternally, but not when either of them is administered independently. On the other hand riluzole, but not FFA, drastically reduced gasping generation and compromised the ability of mice to autoresucitate. Neither substance P nor forskolin was able to reestablish respiratory activity after riluzole and FFA coapplication. Our results confirm in vitro reports and suggest that eupnea generation in neonates requires a complex neuronal network that includes riluzole- and FFA-sensitive elements and that gasping activity depends mostly on a riluzole-sensitive mechanism.
Collapse
Affiliation(s)
- Fernando Peña
- Departamento de Farmacobiología, Cinvestav-Sede Sur, Calz. de los Tenorios 235, Col. Granjas Coapa, 14330 México, D.F., Mexico.
| | | |
Collapse
|
43
|
Cooper EC. Exploiting the other inhibitory ion: KCNQ potassium channels and regulation of excitability in developing and mature brain. Epilepsy Curr 2007; 6:133-5. [PMID: 17260036 PMCID: PMC1783435 DOI: 10.1111/j.1535-7511.2006.00122.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Epileptiform Activity Induced by Pharmacologic Reduction of M-Current in the Developing Hippocampus In Vitro Pena F, Alavez-Perez N Epilepsia 2006;47(1):47–54 Purpose Benign familial neonatal convulsions (BFNCs), an inheritable epilepsy that occurs in neonates but not in adults, is caused by hypofunctional mutations in genes codifying for the M-type K+ current. In an attempt to develop an in vitro model of this disease, we tested whether blocking M-current with linopirdine induces epileptiform activity in brain slices from animals of different ages. Methods Horizontal hippocampus–entorhinal cortex slices were obtained from neonatal (1–2 weeks after birth) and adult (8–9 weeks after birth) rats. Extracellular field recordings of the CA1 region were performed. After recording control conditions, linopirdine was added to the bath, and field activity was recorded continuously for 3 hours. A drug 4-aminopyridine, commonly used to induce epileptiform activity in vitro, was used as a control for our experimental conditions. Results Bath perfusion of linopirdine induced epileptiform activity only in slices from neonatal rats. Epileptiform activity consisted of interictal-like and ictal-like activity. In slices from adult rats, linopirdine induced erratic interictal-like activity. In contrast, 4-aminopyridine was able to induce epileptiform activity in slices from both neonatal and adult rats. Conclusions We demonstrated that blockade of Mcurrent in vitro produces epileptiform activity with a developmental pattern similar to that observed in BNFCs. This could be an in vitro model that can be used to study the cellular mechanisms of epileptogenesis and the developmental features of BFNCs, as well as to develop some therapeutic strategies. Conditional Transgenic Suppression of M Channels in Mouse Brain Reveals Functions in Neuronal Excitability, Resonance and Behavior Peters HC, Hu H, Pongs O, Storm JF, Isbrandt D Nat Neurosci 2005;8(1):51–60 In humans, mutations in the KCNQ2 or KCNQ3 potassium-channel genes are associated with an inherited epilepsy syndrome. We have studied the contribution of KCNQ/M-channels to the control of neuronal excitability by using transgenic mice that conditionally express dominant-negative KCNQ2 subunits in brain. We show that suppression of the neuronal M current in mice is associated with spontaneous seizures, behavioral hyperactivity and morphological changes in the hippocampus. Restriction of transgene expression to defined developmental periods revealed that M-channel activity is critical to the development of normal hippocampal morphology during the first postnatal weeks. Suppression of the M current after this critical period resulted in mice with signs of increased neuronal excitability and deficits in hippocampus-dependent spatial memory. M-current-deficient hippocampal CA1 pyramidal neurons showed increased excitability, reduced spike-frequency adaptation, attenuated medium afterhyperpolarization and reduced intrinsic subthreshold theta resonance. M channels are thus critical determinants of cellular and neuronal network excitability, postnatal brain development and cognitive performance.
Collapse
|
44
|
Abstract
This review considers how recent advances in the physiology of ion channels and other potential molecular targets, in conjunction with new information on the genetics of idiopathic epilepsies, can be applied to the search for improved antiepileptic drugs (AEDs). Marketed AEDs predominantly target voltage-gated cation channels (the alpha subunits of voltage-gated Na+ channels and also T-type voltage-gated Ca2+ channels) or influence GABA-mediated inhibition. Recently, alpha2-delta voltage-gated Ca2+ channel subunits and the SV2A synaptic vesicle protein have been recognized as likely targets. Genetic studies of familial idiopathic epilepsies have identified numerous genes associated with diverse epilepsy syndromes, including genes encoding Na+ channels and GABA(A) receptors, which are known AED targets. A strategy based on genes associated with epilepsy in animal models and humans suggests other potential AED targets, including various voltage-gated Ca2+ channel subunits and auxiliary proteins, A- or M-type voltage-gated K+ channels, and ionotropic glutamate receptors. Recent progress in ion channel research brought about by molecular cloning of the channel subunit proteins and studies in epilepsy models suggest additional targets, including G-protein-coupled receptors, such as GABA(B) and metabotropic glutamate receptors; hyperpolarization-activated cyclic nucleotide-gated cation (HCN) channel subunits, responsible for hyperpolarization-activated current Ih; connexins, which make up gap junctions; and neurotransmitter transporters, particularly plasma membrane and vesicular transporters for GABA and glutamate. New information from the structural characterization of ion channels, along with better understanding of ion channel function, may allow for more selective targeting. For example, Na+ channels underlying persistent Na+ currents or GABA(A) receptor isoforms responsible for tonic (extrasynaptic) currents represent attractive targets. The growing understanding of the pathophysiology of epilepsy and the structural and functional characterization of the molecular targets provide many opportunities to create improved epilepsy therapies.
Collapse
Affiliation(s)
- Brian S Meldrum
- Centre for Neuroscience, Division of Biomedical and Health Sciences, School of Medicine, Kings College, London, United Kingdom
| | | |
Collapse
|
45
|
Lawrence JJ, Saraga F, Churchill JF, Statland JM, Travis KE, Skinner FK, McBain CJ. Somatodendritic Kv7/KCNQ/M channels control interspike interval in hippocampal interneurons. J Neurosci 2006; 26:12325-38. [PMID: 17122058 PMCID: PMC6675427 DOI: 10.1523/jneurosci.3521-06.2006] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The M-current (I(M)), comprised of Kv7 channels, is a voltage-activated K+ conductance that plays a key role in the control of cell excitability. In hippocampal principal cells, I(M) controls action potential (AP) accommodation and contributes to the medium-duration afterhyperpolarization, but the role of I(M) in control of interneuron excitability remains unclear. Here, we investigated I(M) in hippocampal stratum oriens (SO) interneurons, both from wild-type and transgenic mice in which green fluorescent protein (GFP) was expressed in somatostatin-containing interneurons. Somatodendritic expression of Kv7.2 or Kv7.3 subunits was colocalized in a subset of GFP+ SO interneurons, corresponding to oriens-lacunosum moleculare (O-LM) cells. Under voltage clamp (VC) conditions at -30 mV, the Kv7 channel antagonists linopirdine/XE-991 abolished the I(M) amplitude present during relaxation from -30 to -50 mV and reduced the holding current (I(hold)). In addition, 0.5 mM tetraethylammonium reduced I(M), suggesting that I(M) was composed of Kv7.2-containing channels. In contrast, the Kv7 channel opener retigabine increased I(M) amplitude and I(hold). When strongly depolarized in VC, the linopirdine-sensitive outward current activated rapidly and comprised up to 20% of the total current. In current-clamp recordings from GFP+ SO cells, linopirdine induced depolarization and increased AP frequency, whereas retigabine induced hyperpolarization and arrested firing. In multicompartment O-LM interneuron models that incorporated I(M), somatodendritic placement of Kv7 channels best reproduced experimentally measured I(M). The models suggest that Kv3- and Kv7-mediated channels both rapidly activate during single APs; however, Kv3 channels control rapid repolarization of the AP, whereas Kv7 channels primarily control the interspike interval.
Collapse
Affiliation(s)
- J Josh Lawrence
- Laboratory of Cellular and Synaptic Neurophysiology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | | | |
Collapse
|