55 results on '"Hernandez CC"'
Search Results
2. Gating Consequences of Charge Neutralization of Arginine Residues in the S4 Segment of Kv7.2, an Epilepsy-Linked K+ Channel Subunit
- Author
-
Lucio Annunziato, Maria Virginia Soldovieri, Francesco Miceli, Ciria C. Hernandez, Mark S. Shapiro, Maurizio Taglialatela, Miceli, Francesco, Soldovieri, Mv, Hernandez, Cc, Shapiro, M, Annunziato, Lucio, and Taglialatela, Maurizio
- Subjects
Models, Molecular ,Patch-Clamp Techniques ,K+ channel ,Arginine ,Glutamine ,Protein subunit ,Molecular Sequence Data ,Mutant ,Biophysics ,arginine ,CHO Cells ,Gating ,medicine.disease_cause ,03 medical and health sciences ,Cricetulus ,0302 clinical medicine ,Cricetinae ,medicine ,Animals ,Humans ,KCNQ2 Potassium Channel ,Benign familial neonatal seizures ,Amino Acid Sequence ,Channels, Receptors, and Electrical Signaling ,030304 developmental biology ,0303 health sciences ,Mutation ,Chemistry ,Tryptophan ,medicine.disease ,Epilepsy, Benign Neonatal ,Electrophysiology ,Amino Acid Substitution ,Biochemistry ,Mutagenesis, Site-Directed ,epilepsy ,Ion Channel Gating ,030217 neurology & neurosurgery - Abstract
The K(v)7.2 subunits are the main molecular determinants of the M-current, a widespread K(+) current regulating neuronal excitability. Mutations in the K(v)7.2 gene cause benign familial neonatal seizures, an autosomally inherited human epilepsy. The benign familial neonatal seizure-causing mutations include those at arginine residues at positions 207 and 214 in the S(4) segment of K(v)7.2. In this study, each of the six S(4) arginines was individually replaced with neutral glutamines, and the functional properties of mutant channels were studied by whole-cell and single-channel voltage-clamp measurements. The results obtained suggest that each S(4) arginine residue plays a relevant role in the voltage-dependent gating of K(v)7.2 channels. In particular, a decreased positive charge at the N-terminal end of S(4) stabilized the activated state of the voltage-sensor, whereas positive-charge neutralization at the C-terminal end of S(4) favored the resting conformation. Strikingly, neutralization of a single arginine at position 201 was sufficient to cause a significant loss of voltage dependence in channel activation. Moreover, by comparing the functional properties of glutamine versus tryptophan substitution, we found steric bulk to play a relevant role at position 207, but not at position 214, in which the main functional effect of this disease-causing mutation seems to be a consequence of the loss of the positive charge.
- Published
- 2008
- Full Text
- View/download PDF
3. Atypical gating of M-type potassium channels conferred by mutations in uncharged residues in the S4 region of KCNQ2 causing benign familial neonatal convulsions
- Author
-
Maurizio Taglialatela, Lucio Annunziato, Emanuele Miraglia del Giudice, Giulia Bellini, Antonio Pascotto, Francesco Miceli, Pasqualina Castaldo, Maria Virginia Soldovieri, Maria Roberta Cilio, Ciria C. Hernandez, Mark S. Shapiro, Soldovieri, Mv, Cilio, Mr, Miceli, Francesco, Bellini, G, Miraglia del Giudice, E, Castaldo, P, Hernandez, Cc, Shapiro, M, Pascotto, A, Annunziato, Lucio, Taglialatela, Maurizio, Miceli, F, Bellini, Giulia, MIRAGLIA DEL GIUDICE, Emanuele, Pascotto, Antonio, Annunziato, L, Taglialatela, M., and MIRAGLIA DEL GIUDICE, E
- Subjects
Male ,benign familial neonatal convulsion ,Protein subunit ,channel gating ,Molecular Sequence Data ,Gating ,CHO Cells ,medicine.disease_cause ,KCNQ2 subunit ,Membrane Potentials ,Cricetulus ,KCNQ2 Potassium Channel ,Cricetinae ,medicine ,Homomeric ,Animals ,Humans ,Benign familial neonatal seizures ,Amino Acid Sequence ,Membrane potential ,Mutation ,Chemistry ,General Neuroscience ,Infant ,Articles ,medicine.disease ,mutations ,Potassium channel ,Epilepsy, Benign Neonatal ,Pedigree ,Biochemistry ,Amino Acid Substitution ,Child, Preschool ,Biophysics ,epilepsy ,Female ,Ion Channel Gating ,potassium channel - Abstract
Heteromeric assembly of KCNQ2 and KCNQ3 subunits underlie the M-current (IKM), a slowly activating and noninactivating neuronal K+current. Mutations inKCNQ2andKCNQ3genes cause benign familial neonatal convulsions (BFNCs), a rare autosomal-dominant epilepsy of the newborn. In the present study, we describe the identification of a novelKCNQ2heterozygous mutation (c587t) in a BFNC-affected family, leading to an alanine to valine substitution at amino acid position 196 located at the N-terminal end of the voltage-sensing S4domain. The consequences on KCNQ2 subunit function prompted by the A196V substitution, as well as by the A196V/L197P mutation previously described in another BFNC-affected family, were investigated by macroscopic and single-channel current measurements in CHO cells transiently transfected with wild-type and mutant subunits. When compared with KCNQ2 channels, homomeric KCNQ2 A196V or A196V/L197P channels showed a 20 mV rightward shift in their activation voltage dependence, with no concomitant change in maximal open probability or single-channel conductance. Furthermore, current activation kinetics of KCNQ2 A196V channels displayed an unusual dependence on the conditioning prepulse voltage, being markedly slower when preceded by prepulses to more depolarized potentials. Heteromeric channels formed by KCNQ2 A196V and KCNQ3 subunits displayed gating changes similar to those of KCNQ2 A196V homomeric channels. Collectively, these results reveal a novel role for noncharged residues in the N-terminal end of S4in controlling gating ofIKMand suggest that gating changes caused by mutations at these residues may decreaseIKMfunction, thus causing neuronal hyperexcitability, ultimately leading to neonatal convulsions.
- Published
- 2007
4. Clinical characteristics, management, diagnostic findings, and various etiologies of patients with Kounis syndrome. A systematic review.
- Author
-
Cahuapaza-Gutierrez NL, Calderon-Hernandez CC, Chambergo-Michilot D, De Arruda-Chaves E, Zamora A, and Runzer-Colmenares FM
- Subjects
- Humans, Disease Management, Acute Coronary Syndrome diagnosis, Acute Coronary Syndrome epidemiology, Acute Coronary Syndrome therapy, Kounis Syndrome epidemiology, Kounis Syndrome diagnosis, Kounis Syndrome etiology
- Abstract
Background: Kounis syndrome (KS) is defined by the association of acute coronary syndrome secondary to an anaphylactic reaction. KS is often underdiagnosed, and new etiologies have been proposed., Aims: To synthesize the available evidence on clinical profile, management, diagnosis, and etiologies in patients with KS., Methods: A search was conducted in the following databases: PubMed, Scopus, EMBASE and Web of Science from inception to March 19th, 2024. Case reports, case series, and observational studies were included. Letters to the editor, editorials, comments, notes, narrative reviews, and systematic reviews were excluded., Results: A total of 190 studies were included (174 case reports, 13 case series, and 3 observational studies, 214 patients). A predominance of male gender was observed (69.63 %). Mean age was 54.4 ± 16.5 years. The most common comorbidities were hypertension (33.64 %), diabetes (16.82 %), and dyslipidemia (16.35 %). The most frequent clinical manifestations were chest pain (66.35 %) and difficulty breathing (34.11 %). Three variants of KS were identified: type I or allergic coronary vasospasm was the most frequent (43.46 %), and type III, the least common (8.88 %). The most frequent etiology was drug use (38.32 %), primarily antibiotics (42.68 %), followed by animal stings or bites (26.17 %). The calculated KS rate was 11.12 per 1000 people. The mortality rate was 7.47 %, and the majority had a favorable outcome (86.92 %) after management., Conclusions: KS is a complex and underdiagnosed disease that should be considered as a differential diagnosis in acute coronary syndrome associated with an allergic reaction., Competing Interests: Declaration of competing interest The authors declare that they have no conflict of interests., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
5. N -(4-Bromo-2,5-Dimethoxyphenethyl)-6-(4-Phenylbutoxy)Hexan-1-Amine (XOB): A Novel Phenylalkylamine Antagonist of Serotonin 2A Receptors and Voltage-Gated Sodium Channels.
- Author
-
Denomme N, Hernandez CC, Bock HA, Ohana RF, Bakshi S, Sherwood AM, McCorvy JD, Daley PF, Callaway WB, Hull JM, Alt A, Isom LL, and Cozzi NV
- Subjects
- Animals, Mice, Humans, Serotonin 5-HT2 Receptor Antagonists pharmacology, Voltage-Gated Sodium Channels metabolism, Voltage-Gated Sodium Channels drug effects, Male, Mice, Inbred C57BL, HEK293 Cells, Cricetulus, Receptor, Serotonin, 5-HT2A metabolism, Receptor, Serotonin, 5-HT2A drug effects
- Abstract
Bipolar disorder impacts millions of patients in the United States but the mechanistic understanding of its pathophysiology and therapeutics is incomplete. Atypical antipsychotic serotonin
2A (5-HT2A ) receptor antagonists, such as quetiapine and olanzapine, and mood-stabilizing voltage-gated sodium channel (VGSC) blockers, such as lamotrigine, carbamazepine, and valproate, show therapeutic synergy and are often prescribed in combination for the treatment of bipolar disorder. Combination therapy is a complex task for clinicians and patients, often resulting in unexpected difficulties with dosing, drug tolerances, and decreased patient compliance. Thus, an unmet need for bipolar disorder treatment is to develop a therapeutic agent that targets both 5-HT2A receptors and VGSCs. Toward this goal, we developed a novel small molecule that simultaneously antagonizes 5-HT2A receptors and blocks sodium current. The new compound, N -(4-bromo-2,5-dimethoxyphenethyl)-6-(4-phenylbutoxy)hexan-1-amine (XOB) antagonizes 5-HT-stimulated, Gq -mediated, calcium flux at 5-HT2A receptors at low micromolar concentrations while displaying negligible affinity and activity at 5-HT1A , 5-HT2B , and 5-HT2C receptors. At similar concentrations, XOB administration inhibits sodium current in heterologous cells and results in reduced action potential (AP) firing and VGSC-related AP properties in mouse prefrontal cortex layer V pyramidal neurons. Thus, XOB represents a new, proof-of-principle tool that can be used for future preclinical investigations and therapeutic development. This polypharmacology approach of developing a single molecule to act upon two targets, which are currently independently targeted by combination therapies, may lead to safer alternatives for the treatment of psychiatric disorders that are increasingly being found to benefit from the simultaneous targeting of multiple receptors. SIGNIFICANCE STATEMENT: The authors synthesized a novel small molecule (XOB) that simultaneously antagonizes two key therapeutic targets of bipolar disorder, 5-HT2A receptors and voltage-gated sodium channels, in heterologous cells, and inhibits the intrinsic excitability of mouse prefrontal cortex layer V pyramidal neurons in brain slices. XOB represents a valuable new proof-of-principle tool for future preclinical investigations and provides a novel molecular approach to the pharmacological treatment of complex neuropsychiatric disease, which often requires a combination of therapeutics for sufficient patient benefit., (Copyright © 2024 by The American Society for Pharmacology and Experimental Therapeutics.)- Published
- 2024
- Full Text
- View/download PDF
6. Functional coupling between MC4R and Kir7.1 contributes to clozapine-induced hyperphagia.
- Author
-
Li L, Hernandez CC, Gimenez LE, Xu B, Dahir NS, Swati, Birnbaum SG, Cone RD, and Liu C
- Abstract
Most antipsychotic drugs (APDs) induce hyperphagia and weight gain. However, the neural mechanisms are poorly understood, partly due to challenges replicating their metabolic effects in rodents. Here, we report a new mouse model that recapitulates overeating induced by clozapine, a widely prescribed APD. Our study shows that clozapine boosts food intake by inhibiting melanocortin 4 receptor (MC4R) expressing neurons in the paraventricular nucleus of the hypothalamus. Interestingly, neither clozapine nor risperidone, another commonly used APD, affects receptor-ligand binding or the canonical Gαs signaling of MC4R. Instead, they inhibit neuronal activity by enhancing the coupling between MC4R and Kir7.1, leading to the open state of the inwardly rectifying potassium channel. Deletion of Kir7.1 in Mc4r-Cre neurons prevents clozapine-induced weight gain, while treatment with a selective Kir7.1 blocker mitigates overeating in clozapine-fed mice. Our findings unveil a molecular pathway underlying the effect of APDs on feeding behavior and suggest its potential as a therapeutic target.
- Published
- 2024
- Full Text
- View/download PDF
7. Structure of the Ion Channel Kir7.1 and Implications for its Function in Normal and Pathophysiologic States.
- Author
-
Peisley A, Hernandez CC, Dahir NS, Koepping L, Raczkowski A, Su M, Ghamari-Langroudi M, Ji X, Gimenez LE, and Cone RD
- Abstract
Hereditary defects in the function of the Kir7.1 in the retinal pigment epithelium are associated with the ocular diseases retinitis pigmentosa, Leber congenital amaurosis, and snowflake vitreal degeneration. Studies also suggest that Kir7.1 may be regulated by a GPCR, the melanocortin-4 receptor, in certain hypothalamic neurons. We present the first structures of human Kir7.1 and describe the conformational bias displayed by two pathogenic mutations, R162Q and E276A, to provide an explanation for the basis of disease and illuminate the gating pathway. We also demonstrate the structural basis for the blockade of the channel by a small molecule ML418 and demonstrate that channel blockade in vivo activates MC4R neurons in the paraventricular nucleus of the hypothalamus (PVH), inhibiting food intake and inducing weight loss. Preliminary purification, and structural and pharmacological characterization of an in tandem construct of MC4R and Kir7.1 suggests that the fusion protein forms a homotetrameric channel that retains regulation by liganded MC4R molecules., Competing Interests: DECLARATION OF INTERESTS RDC, LEG, and the University of Michigan have equity in Courage Therapeutics, and RDC serves on the company board. The other authors declare no competing interests.
- Published
- 2024
- Full Text
- View/download PDF
8. Editorial: Targeting ion channels for drug discovery: emerging challenges for high throughput screening technologies.
- Author
-
Hernandez CC, Gimenez LE, Strassmaier T, Rogers M, and Taymans JM
- Abstract
Competing Interests: LG and the University of Michigan have equity in Courage Therapeutics. TS was employed by Nanion Technologies Inc. and MR was employed by Albion Drug Discovery Services Ltd. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision.
- Published
- 2024
- Full Text
- View/download PDF
9. Anti-diabetic and anti-inflammatory bioactive hits from Coriaria intermedia Matsum. stem and Dracontomelon dao (Blanco) Merr. & Rolfe bark through bioassay-guided fractionation and liquid chromatography-tandem mass spectrometry.
- Author
-
Fabian MCP, Astorga RMN, Atis AAG, Pilapil LAE, and Hernandez CC
- Abstract
Women have been found to be at a higher risk of morbidity and mortality from type 2 diabetes mellitus (T2DM) and asthma. α-Glucosidase inhibitors have been used to treat T2DM, and arachidonic acid 15-lipoxygenase (ALOX15) inhibitors have been suggested to be used as treatments for asthma and T2DM. Compounds that inhibit both enzymes may be studied as potential treatments for people with both T2DM and asthma. This study aimed to determine potential anti-diabetic and anti-inflammatory bioactive hits from Coriaria intermedia Matsum. stem and Dracontomelon dao (Blanco) Merr. & Rolfe bark. A bioassay-guided fractionation framework was used to generate bioactive fractions from C. intermedia stem and D. dao bark. Subsequently, dereplication through ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) and database searching was performed to putatively identify the components of one bioactive fraction from each plant. Seven compounds were putatively identified from the C. intermedia stem active fraction, and six of these compounds were putatively identified from this plant for the first time. Nine compounds were putatively identified from the D. dao bark active fraction, and seven of these compounds were putatively identified from this plant for the first time. One putative compound from the C. intermedia stem active fraction (corilagin) has been previously reported to have inhibitory activity against both α-glucosidase and 15-lipoxygenase-1. It is suggested that further studies on the potential of corilagin as an anti-diabetic and anti-inflammatory treatment should be pursued based on its several beneficial pharmacological activities and its low reported toxicity., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Fabian, Astorga, Atis, Pilapil and Hernandez.)
- Published
- 2024
- Full Text
- View/download PDF
10. Novel Cocrystal Structures of Peptide Antagonists Bound to the Human Melanocortin Receptor 4 Unveil Unexplored Grounds for Structure-Based Drug Design.
- Author
-
Gimenez LE, Martin C, Yu J, Hollanders C, Hernandez CC, Wu Y, Yao D, Han GW, Dahir NS, Wu L, Van der Poorten O, Lamouroux A, Mannes M, Zhao S, Tourwé D, Stevens RC, Cone RD, and Ballet S
- Subjects
- Humans, Ligands, Drug Design, Receptor, Melanocortin, Type 3, Receptors, Melanocortin, Receptor, Melanocortin, Type 4, Peptides pharmacology
- Abstract
Melanocortin 4 receptor (MC4-R) antagonists are actively sought for treating cancer cachexia. We determined the structures of complexes with PG-934 and SBL-MC-31 . These peptides differ from SHU9119 by substituting His
6 with Pro6 and inserting Gly10 or Arg10 . The structures revealed two subpockets at the TM7-TM1-TM2 domains, separated by N2857.36 . Two peptide series based on the complexed peptides led to an antagonist activity and selectivity SAR study. Most ligands retained the SHU9119 potency, but several SBL-MC-31 -derived peptides significantly enhanced MC4-R selectivity over MC1-R by 60- to 132-fold. We also investigated MC4-R coupling to the K+ channel, Kir7.1. Some peptides activated the channel, whereas others induced channel closure independently of G protein coupling. In cell culture studies, channel activation correlated with increased feeding, while a peptide with Kir7.1 inhibitory activity reduced eating. These results highlight the potential for targeting the MC4-R:Kir7.1 complex for treating positive and restrictive eating disorders.- Published
- 2024
- Full Text
- View/download PDF
11. Automated Patch Clamp Recordings of GPCR-Gated Ion Channels: Targeting the MC4-R/Kir7.1 Potassium Channel Complex.
- Author
-
Hernandez CC, Gimenez LE, and Cone RD
- Subjects
- Animals, Humans, Ion Channel Gating drug effects, Receptors, G-Protein-Coupled metabolism, HEK293 Cells, Patch-Clamp Techniques methods, Receptor, Melanocortin, Type 4 metabolism, Potassium Channels, Inwardly Rectifying metabolism
- Abstract
Automated patch clamp recording is a valuable technique in drug discovery and the study of ion channels. It allows for the precise measurement and manipulation of channel currents, providing insights into their function and modulation by drugs or other compounds. The melanocortin 4 receptor (MC4-R) is a G protein-coupled receptor (GPCR) crucial to appetite regulation, energy balance, and body weight. MC4-R signaling is complex and involves interactions with other receptors and neuropeptides in the appetite-regulating circuitry. MC4-Rs, like other GPCRs, are known to modulate ion channels such as Kir7.1, an inward rectifier potassium channel, in response to ligand binding. This modulation is critical for controlling ion flow across the cell membrane, which can influence membrane potential, excitability, and neurotransmission. The MC4-R is the target for the anti-obesity drug Imcivree. However, this drug is known to lack optimal potency and also has side effects. Using high-throughput techniques for studying the MC4-R/Kir7.1 complex allows researchers to rapidly screen many compounds or conditions, aiding the development of drugs that target this system. Additionally, automated patch clamp recording of this receptor-channel complex and its ligands can provide valuable functional and pharmacological insights supporting the development of novel therapeutic strategies. This approach can be generalized to other GPCR-gated ion channel functional complexes, potentially accelerating the pace of research in different fields with the promise to uncover previously unknown aspects of receptor-ion channel interactions., (© 2024. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
12. Epileptic Encephalopathy GABRB Structural Variants Share Common Gating and Trafficking Defects.
- Author
-
Hernandez CC, Hu N, Shen W, and Macdonald RL
- Subjects
- Humans, Mutation, Missense, Mutation, Seizures, Receptors, GABA-A metabolism, Brain Diseases
- Abstract
Variants in the GABRB gene, which encodes the β subunit of the GABA
A receptor, have been implicated in various epileptic encephalopathies and related neurodevelopmental disorders such as Dravet syndrome and Angelman syndrome. These conditions are often associated with early-onset seizures, developmental regression, and cognitive impairments. The severity and specific features of these encephalopathies can differ based on the nature of the genetic variant and its impact on GABAA receptor function. These variants can lead to dysfunction in GABAA receptor-mediated inhibition, resulting in an imbalance between neuronal excitation and inhibition that contributes to the development of seizures. Here, 13 de novo EE-associated GABRB variants, occurring as missense mutations, were analyzed to determine their impact on protein stability and flexibility, channel function, and receptor biogenesis. Our results showed that all mutations studied significantly impact the protein structure, altering protein stability, flexibility, and function to varying degrees. Variants mapped to the GABA-binding domain, coupling zone, and pore domain significantly impact the protein structure, modifying the β+/α- interface of the receptor and altering channel activation and receptor trafficking. Our study proposes that the extent of loss or gain of GABAA receptor function can be elucidated by identifying the specific structural domain impacted by mutation and assessing the variability in receptor structural dynamics. This paves the way for future studies to explore and uncover links between the incidence of a variant in the receptor topology and the severity of the related disease.- Published
- 2023
- Full Text
- View/download PDF
13. Targeting the central melanocortin system for the treatment of metabolic disorders.
- Author
-
Sweeney P, Gimenez LE, Hernandez CC, and Cone RD
- Subjects
- Humans, Melanocortins therapeutic use, Obesity drug therapy, Cachexia, Sexual Dysfunctions, Psychological drug therapy, Sexual Dysfunctions, Psychological metabolism, Metabolic Diseases drug therapy
- Abstract
A large body of preclinical and clinical data shows that the central melanocortin system is a promising therapeutic target for treating various metabolic disorders such as obesity and cachexia, as well as anorexia nervosa. Setmelanotide, which functions by engaging the central melanocortin circuitry, was approved by the FDA in 2020 for use in certain forms of syndromic obesity. Furthermore, the FDA approvals in 2019 of two peptide drugs targeting melanocortin receptors for the treatment of generalized hypoactive sexual desire disorder (bremelanotide) and erythropoietic protoporphyria-associated phototoxicity (afamelanotide) demonstrate the safety of this class of peptides. These approvals have also renewed excitement in the development of therapeutics targeting the melanocortin system. Here, we review the anatomy and function of the melanocortin system, discuss progress and challenges in developing melanocortin receptor-based therapeutics, and outline potential metabolic and behavioural disorders that could be addressed using pharmacological agents targeting these receptors., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
14. The unique structural characteristics of the Kir 7.1 inward rectifier potassium channel: a novel player in energy homeostasis control.
- Author
-
Hernandez CC, Gimenez LE, Dahir NS, Peisley A, and Cone RD
- Subjects
- Humans, Mutation, Neurons metabolism, Potassium metabolism, Protein Domains, Potassium Channels, Inwardly Rectifying genetics, Potassium Channels, Inwardly Rectifying metabolism
- Abstract
The inward rectifier potassium channel Kir7.1, encoded by the KCNJ13 gene, is a tetramer composed of two-transmembrane domain-spanning monomers, closer in homology to Kir channels associated with potassium transport such as Kir1.1, 1.2, and 1.3. Compared with other channels, Kir7.1 exhibits small unitary conductance and low dependence on external potassium. Kir7.1 channels also show a phosphatidylinositol 4,5-bisphosphate (PIP
2 ) dependence for opening. Accordingly, retinopathy-associated Kir7.1 mutations mapped at the binding site for PIP2 resulted in channel gating defects leading to channelopathies such as snowflake vitreoretinal degeneration and Leber congenital amaurosis in blind patients. Lately, this channel's role in energy homeostasis was reported due to the direct interaction with the melanocortin type 4 receptor (MC4R) in the hypothalamus. As this channel seems to play a multipronged role in potassium homeostasis and neuronal excitability, we will discuss what is predicted from a structural viewpoint and its possible implications for hunger control.- Published
- 2023
- Full Text
- View/download PDF
15. GABRG2 Variants Associated with Febrile Seizures.
- Author
-
Hernandez CC, Shen Y, Hu N, Shen W, Narayanan V, Ramsey K, He W, Zou L, and Macdonald RL
- Subjects
- Humans, Receptors, GABA-A genetics, Mutation, Missense, Mutation, Seizures, Febrile genetics, Epilepsies, Myoclonic genetics, Epilepsy genetics
- Abstract
Febrile seizures (FS) are the most common form of epilepsy in children between six months and five years of age. FS is a self-limited type of fever-related seizure. However, complicated prolonged FS can lead to complex partial epilepsy. We found that among the GABA
A receptor subunit ( GABR ) genes, most variants associated with FS are harbored in the γ2 subunit ( GABRG2 ). Here, we characterized the effects of eight variants in the GABAA receptor γ2 subunit on receptor biogenesis and channel function. Two-thirds of the GABRG2 variants followed the expected autosomal dominant inheritance in FS and occurred as missense and nonsense variants. The remaining one-third appeared as de novo in the affected probands and occurred only as missense variants. The loss of GABAA receptor function and dominant negative effect on GABAA receptor biogenesis likely caused the FS phenotype. In general, variants in the GABRG2 result in a broad spectrum of phenotypic severity, ranging from asymptomatic, FS, genetic epilepsy with febrile seizures plus (GEFS+), and Dravet syndrome individuals. The data presented here support the link between FS, epilepsy, and GABRG2 variants, shedding light on the relationship between the variant topological occurrence and disease severity.- Published
- 2023
- Full Text
- View/download PDF
16. Development of an automated screen for Kv7.2 potassium channels and discovery of a new agonist chemotype.
- Author
-
Hernandez CC, Tarfa RA, Miguel I Limcaoco J, Liu R, Mondal P, Hill C, Keith Duncan R, Tzounopoulos T, Stephenson CRJ, O'Meara MJ, and Wipf P
- Subjects
- KCNQ Potassium Channels genetics, KCNQ Potassium Channels metabolism, KCNQ2 Potassium Channel genetics, KCNQ2 Potassium Channel metabolism
- Abstract
To identify pore domain ligands on Kv7.2 potassium ion channels, we compared wild-type (WT) and W236L mutant Kv7.2 channels in a series of assays with previously validated and novel agonist chemotypes. Positive controls were retigabine, flupirtine, and RL-81; i.e. Kv7.2 channel activators that significantly shift voltage-dependent activation to more negative potentials (ΔV
50 ) at 5 µM. We identified 6 new compounds that exhibited differential enhancing activity between WT and W236L mutant channels. Whole cell patch-clamp electrophysiology studies were conducted to identify Kv7.2. Kv7.2/3, Kv7.4, and Kv7.5 selectivity. Our results validate the SyncroPatch platform and establish new structure activity relationships (SAR). Specifically, in addition to selective Kv7.2, Kv7.2/3, Kv7.4. and Kv7.5 agonists, we identified a novel chemotype, ZK-21, a 4-aminotetrahydroquinoline that is distinct from any of the previously described Kv7 channel modifiers. Using flexible receptor docking, ZK-21 was predicted to be stabilized by W236 and bind perpendicular to retigabine, burying the benzyl carbamate group into a tunnel reaching the core of the pore domain., (Copyright © 2022. Published by Elsevier Ltd.)- Published
- 2022
- Full Text
- View/download PDF
17. An Adaptive Imitation Learning Framework for Robotic Complex Contact-Rich Insertion Tasks.
- Author
-
Wang Y, Beltran-Hernandez CC, Wan W, and Harada K
- Abstract
Complex contact-rich insertion is a ubiquitous robotic manipulation skill and usually involves nonlinear and low-clearance insertion trajectories as well as varying force requirements. A hybrid trajectory and force learning framework can be utilized to generate high-quality trajectories by imitation learning and find suitable force control policies efficiently by reinforcement learning. However, with the mentioned approach, many human demonstrations are necessary to learn several tasks even when those tasks require topologically similar trajectories. Therefore, to reduce human repetitive teaching efforts for new tasks, we present an adaptive imitation framework for robot manipulation. The main contribution of this work is the development of a framework that introduces dynamic movement primitives into a hybrid trajectory and force learning framework to learn a specific class of complex contact-rich insertion tasks based on the trajectory profile of a single task instance belonging to the task class. Through experimental evaluations, we validate that the proposed framework is sample efficient, safer, and generalizes better at learning complex contact-rich insertion tasks on both simulation environments and on real hardware., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Wang, Beltran-Hernandez, Wan and Harada.)
- Published
- 2022
- Full Text
- View/download PDF
18. Dravet syndrome-associated mutations in GABRA1 , GABRB2 and GABRG2 define the genetic landscape of defects of GABA A receptors.
- Author
-
Hernandez CC, Tian X, Hu N, Shen W, Catron MA, Yang Y, Chen J, Jiang Y, Zhang Y, and Macdonald RL
- Abstract
Dravet syndrome is a rare, catastrophic epileptic encephalopathy that begins in the first year of life, usually with febrile or afebrile hemiclonic or generalized tonic-clonic seizures followed by status epilepticus. De novo variants in genes that mediate synaptic transmission such as SCN1A and PCDH19 are often associated with Dravet syndrome. Recently, GABA
A receptor subunit genes ( GABRs ) encoding α1 ( GABRA1 ), β3 ( GABRB3 ) and γ2 ( GABRG2 ), but not β2 ( GABRB2 ) or β1 ( GABRB1 ), subunits are frequently associated with Dravet syndrome or Dravet syndrome-like phenotype. We performed next generation sequencing on 870 patients with Dravet syndrome and identified nine variants in three different GABRs . Interestingly, the variants were all in genes encoding the most common GABAA receptor, the α1β2γ2 receptor. Mutations in GABRA1 (c.644T>C, p. L215P; c.640C>T, p. R214C; c.859G>A; V287I; c.641G>A, p. R214H) and GABRG2 (c.269C>G, p. T90R; c.1025C>T, p. P342L) presented as de novo cases, while in GABRB2 two variants were de novo (c.992T>C, p. F331S; c.542A>T, p. Y181F) and one was autosomal dominant and inherited from the maternal side (c.990_992del, p.330_331del). We characterized the effects of these GABR variants on GABAA receptor biogenesis and channel function. We found that defects in receptor gating were the common deficiency of GABRA1 and GABRB2 Dravet syndrome variants, while mainly trafficking defects were found with the GABRG2 (c.269C>G, p. T90R) variant. It seems that variants in α1 and β2 subunits are less tolerated than in γ2 subunits, since variant α1 and β2 subunits express well but were functionally deficient. This suggests that all of these GABR variants are all targeting GABR genes that encode the assembled α1β2γ2 receptor, and regardless of which of the three subunits are mutated, variants in genes coding for α1, β2 and γ2 receptor subunits make them candidate causative genes in the pathogenesis of Dravet syndrome., (© The Author(s) (2021). Published by Oxford University Press on behalf of the Guarantors of Brain.)- Published
- 2021
- Full Text
- View/download PDF
19. Membrane orientation and oligomerization of the melanocortin receptor accessory protein 2.
- Author
-
Chen V, Bruno AE, Britt LL, Hernandez CC, Gimenez LE, Peisley A, Cone RD, and Millhauser GL
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Cell Membrane genetics, HEK293 Cells, Humans, Protein Domains, Adaptor Proteins, Signal Transducing metabolism, Cell Membrane metabolism, Protein Multimerization
- Abstract
The melanocortin receptor accessory protein 2 (MRAP2) plays a pivotal role in the regulation of several G protein-coupled receptors that are essential for energy balance and food intake. MRAP2 loss-of-function results in obesity in mammals. MRAP2 and its homolog MRAP1 have an unusual membrane topology and are the only known eukaryotic proteins that thread into the membrane in both orientations. In this study, we demonstrate that the conserved polybasic motif that dictates the membrane topology and dimerization of MRAP1 does not control the membrane orientation and dimerization of MRAP2. We also show that MRAP2 dimerizes through its transmembrane domain and can form higher-order oligomers that arrange MRAP2 monomers in a parallel orientation. Investigating the molecular details of MRAP2 structure is essential for understanding the mechanism by which it regulates G protein-coupled receptors and will aid in elucidating the pathways involved in metabolic dysfunction., Competing Interests: Conflict of interest—The authors declare that they have no conflicts of interest with the contents of this article., (© 2020 Chen et al.)
- Published
- 2020
- Full Text
- View/download PDF
20. Determination of the melanocortin-4 receptor structure identifies Ca 2+ as a cofactor for ligand binding.
- Author
-
Yu J, Gimenez LE, Hernandez CC, Wu Y, Wein AH, Han GW, McClary K, Mittal SR, Burdsall K, Stauch B, Wu L, Stevens SN, Peisley A, Williams SY, Chen V, Millhauser GL, Zhao S, Cone RD, and Stevens RC
- Subjects
- Crystallography, X-Ray, Cyclic AMP chemistry, Humans, Ligands, Melanocyte-Stimulating Hormones chemistry, Melanocyte-Stimulating Hormones pharmacology, Mutation, Potassium Channels, Inwardly Rectifying chemistry, Protein Binding, Protein Multimerization, Protein Structure, Secondary, Receptor, Melanocortin, Type 4 antagonists & inhibitors, Receptor, Melanocortin, Type 4 genetics, Receptors, G-Protein-Coupled antagonists & inhibitors, Receptors, G-Protein-Coupled genetics, Signal Transduction, Calcium chemistry, Receptor, Melanocortin, Type 4 chemistry, Receptors, G-Protein-Coupled chemistry
- Abstract
The melanocortin-4 receptor (MC4R) is involved in energy homeostasis and is an important drug target for syndromic obesity. We report the structure of the antagonist SHU9119-bound human MC4R at 2.8-angstrom resolution. Ca
2+ is identified as a cofactor that is complexed with residues from both the receptor and peptide ligand. Extracellular Ca2+ increases the affinity and potency of the endogenous agonist α-melanocyte-stimulating hormone at the MC4R by 37- and 600-fold, respectively. The ability of the MC4R crystallized construct to couple to ion channel Kir7.1, while lacking cyclic adenosine monophosphate stimulation, highlights a heterotrimeric GTP-binding protein (G protein)-independent mechanism for this signaling modality. MC4R is revealed as a structurally divergent G protein-coupled receptor (GPCR), with more similarity to lipidic GPCRs than to the homologous peptidic GPCRs., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2020
- Full Text
- View/download PDF
21. The GPCR accessory protein MRAP2 regulates both biased signaling and constitutive activity of the ghrelin receptor GHSR1a.
- Author
-
Rouault AAJ, Rosselli-Murai LK, Hernandez CC, Gimenez LE, Tall GG, and Sebag JA
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Animals, CHO Cells, Cricetulus, GTP-Binding Protein alpha Subunits, Gq-G11 genetics, GTP-Binding Protein alpha Subunits, Gq-G11 metabolism, Humans, Receptors, Ghrelin genetics, Adaptor Proteins, Signal Transducing metabolism, Energy Metabolism, Receptors, Ghrelin immunology, Signal Transduction
- Abstract
Ghrelin is a hormone secreted by the stomach during fasting periods and acts through its receptor, the growth hormone secretagogue 1a (GHSR1a), to promote food intake and prevent hypoglycemia. As such, GHSR1a is an important regulator of energy and glucose homeostasis and a target for the treatment of obesity. Here, we showed that the accessory protein MRAP2 altered GHSR1a signaling by inhibiting its constitutive activity, as well as by enhancing its G protein-dependent signaling and blocking the recruitment and signaling of β-arrestin in response to ghrelin. In addition, the effects of MRAP2 on the Gα
q and β-arrestin pathways were independent and involved distinct regions of MRAP2. These findings may have implications for the regulation of ghrelin function in vivo and the role of MRAP2 in energy homeostasis. They also show that accessory proteins can bias signaling downstream of GPCRs in response to their endogenous agonist., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2020
- Full Text
- View/download PDF
22. Altered inhibitory synapses in de novo GABRA5 and GABRA1 mutations associated with early onset epileptic encephalopathies.
- Author
-
Hernandez CC, XiangWei W, Hu N, Shen D, Shen W, Lagrange AH, Zhang Y, Dai L, Ding C, Sun Z, Hu J, Zhu H, Jiang Y, and Macdonald RL
- Subjects
- Child, Child, Preschool, Epilepsy complications, Female, Genetic Predisposition to Disease genetics, Humans, Intellectual Disability complications, Male, Membrane Potentials physiology, Miniature Postsynaptic Potentials physiology, Mutation, Primary Cell Culture, Receptors, GABA-A biosynthesis, Receptors, GABA-A metabolism, Receptors, GABA-A physiology, Synapses physiology, Young Adult, gamma-Aminobutyric Acid metabolism, Epilepsy genetics, Intellectual Disability genetics, Receptors, GABA-A genetics, Synapses genetics
- Abstract
We performed next generation sequencing on 1696 patients with epilepsy and intellectual disability using a gene panel with 480 epilepsy-related genes including all GABAA receptor subunit genes (GABRs), and we identified six de novo GABR mutations, two novel GABRA5 mutations (c.880G>T, p.V294F and c.1238C>T, p.S413F), two novel GABRA1 mutations (c.778C>T, p.P260S and c.887T>C, p.L296S/c.944G>T, p.W315L) and two known GABRA1 mutations (c.335G>A, p.R112Q and c.343A>G, p.N115D) in six patients with intractable early onset epileptic encephalopathy. The α5(V294F and S413F) and α1(P260S and L296S/W315L) subunit residue substitutions were all in transmembrane domains, while the α1(R112Q and N115R) subunit residue substitutions were in the N-terminal GABA binding domain. Using multidisciplinary approaches, we compared effects of mutant GABAA receptor α5 and α1 subunits on the properties of recombinant α5β3γ2 and α1β3γ2 GABAA receptors in both neuronal and non-neuronal cells and characterized their effects on receptor clustering, biogenesis and channel function. GABAA receptors containing mutant α5 and α1 subunits all had reduced cell surface and total cell expression with altered endoplasmic reticulum processing, impaired synaptic clustering, reduced GABAA receptor function and decreased GABA binding potency. Our study identified GABRA5 as a causative gene for early onset epileptic encephalopathy and expands the mutant GABRA1 phenotypic spectrum, supporting growing evidence that defects in GABAergic neurotransmission contribute to early onset epileptic encephalopathy phenotypes., (© The Author(s) (2019). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
23. A structural look at GABA A receptor mutations linked to epilepsy syndromes.
- Author
-
Hernandez CC and Macdonald RL
- Subjects
- Epilepsy genetics, Epilepsy physiopathology, Epileptic Syndromes metabolism, Epileptic Syndromes physiopathology, Humans, Mutation genetics, Receptors, GABA-A physiology, Structure-Activity Relationship, Epileptic Syndromes genetics, Receptors, GABA-A genetics, Receptors, GABA-A metabolism
- Abstract
Understanding the genetic variation in GABA
A receptor subunit genes (GABRs), GABRA1-6, GABRB1-3, GABRG1-3 and GABRD, in individuals affected by epilepsy may improve the diagnosis and treatment of epilepsy syndromes through identification of disease-associated variants. However, the lack of functional analysis and validation of many novel and previously reported familial and de novo mutations have made it challenging to address meaningful gene associations with epilepsy syndromes. GABAA receptors belong to the Cys-loop receptor family. Even though GABAA receptor mutant residues are widespread among different GABRs, their frequent occurrence in important structural domains that share common functional features suggests associations between structure and function., (Copyright © 2019 Elsevier B.V. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
24. Phosphatidylinositol 4,5-bisphosphate (PIP 2 ) regulates KCNQ3 K + channels by interacting with four cytoplasmic channel domains.
- Author
-
Choveau FS, De la Rosa V, Bierbower SM, Hernandez CC, and Shapiro MS
- Subjects
- Amino Acid Sequence, Animals, CHO Cells, Cricetulus, Humans, KCNQ3 Potassium Channel genetics, Models, Molecular, Mutation, Protein Binding, Protein Conformation, alpha-Helical, Protein Domains, Cytoplasm metabolism, KCNQ3 Potassium Channel chemistry, KCNQ3 Potassium Channel metabolism, Phosphatidylinositol 4,5-Diphosphate metabolism
- Abstract
Phosphatidylinositol 4,5-bisphosphate (PIP
2 ) in the plasma membrane regulates the function of many ion channels, including M-type (potassium voltage-gated channel subfamily Q member (KCNQ), Kv 7) K+ channels; however, the molecular mechanisms involved remain unclear. To this end, we here focused on the KCNQ3 subtype that has the highest apparent affinity for PIP2 and performed extensive mutagenesis in regions suggested to be involved in PIP2 interactions among the KCNQ family. Using perforated patch-clamp recordings of heterologously transfected tissue culture cells, total internal reflection fluorescence microscopy, and the zebrafish ( Danio rerio ) voltage-sensitive phosphatase to deplete PIP2 as a probe, we found that PIP2 regulates KCNQ3 channels through four different domains: 1) the A-B helix linker that we previously identified as important for both KCNQ2 and KCNQ3, 2) the junction between S6 and the A helix, 3) the S2-S3 linker, and 4) the S4-S5 linker. We also found that the apparent strength of PIP2 interactions within any of these domains was not coupled to the voltage dependence of channel activation. Extensive homology modeling and docking simulations with the WT or mutant KCNQ3 channels and PIP2 were consistent with the experimental data. Our results indicate that PIP2 modulates KCNQ3 channel function by interacting synergistically with a minimum of four cytoplasmic domains., (© 2018 Choveau et al.)- Published
- 2018
- Full Text
- View/download PDF
25. G protein-coupled receptors differentially regulate glycosylation and activity of the inwardly rectifying potassium channel Kir7.1.
- Author
-
Carrington SJ, Hernandez CC, Swale DR, Aluko OA, Denton JS, and Cone RD
- Subjects
- Glycosylation, HEK293 Cells, Humans, Ion Channel Gating physiology, Leber Congenital Amaurosis genetics, Mutation, Potassium Channels, Inwardly Rectifying chemistry, Potassium Channels, Inwardly Rectifying genetics, Protein Multimerization physiology, Protein Transport physiology, Receptors, Adrenergic, beta-1 metabolism, Receptors, Adrenergic, beta-3 metabolism, Sequence Deletion, Potassium Channels, Inwardly Rectifying metabolism, Receptors, Adrenergic, beta-2 metabolism
- Abstract
Kir7.1 is an inwardly rectifying potassium channel with important roles in the regulation of the membrane potential in retinal pigment epithelium, uterine smooth muscle, and hypothalamic neurons. Regulation of G protein-coupled inwardly rectifying potassium (GIRK) channels by G protein-coupled receptors (GPCRs) via the G protein βγ subunits has been well characterized. However, how Kir channels are regulated is incompletely understood. We report here that Kir7.1 is also regulated by GPCRs, but through a different mechanism. Using Western blotting analysis, we observed that multiple GPCRs tested caused a striking reduction in the complex glycosylation of Kir7.1. Further, GPCR-mediated reduction of Kir7.1 glycosylation in HEK293T cells did not alter its expression at the cell surface but decreased channel activity. Of note, mutagenesis of the sole Kir7.1 glycosylation site reduced conductance and open probability, as indicated by single-channel recording. Additionally, we report that the L241P mutation of Kir7.1 associated with Lebers congenital amaurosis (LCA), an inherited retinal degenerative disease, has significantly reduced complex glycosylation. Collectively, these results suggest that Kir7.1 channel glycosylation is essential for function, and this activity within cells is suppressed by most GPCRs. The melanocortin-4 receptor (MC4R), a GPCR previously reported to induce ligand-regulated activity of this channel, is the only GPCR tested that does not have this effect on Kir7.1., (© 2018 Carrington et al.)
- Published
- 2018
- Full Text
- View/download PDF
26. X-ray powder diffraction and other analyses of cellulose nanocrystals obtained from corn straw by chemical treatments.
- Author
-
Hernandez CC, Ferreira FF, and Rosa DS
- Abstract
The extraction of nanocellulose from agro-industrial wastes is feasible due to a significant amount of cellulose contained in these natural fibers. The analysis of chemical treatments effects on the fibers to obtain the nanocellulose must be taken into consideration for the definition of an adequate and efficient methodology. In this study, two alkaline treatments were used (cleaning and bleaching), as well as an acid treatment for the extraction of nanocellulose from corn straw residues. The samples were characterized using Fourier transform infrared spectroscopy (FTIR), X-ray powder diffraction (XRPD), scanning electron microscopy (SEM), dynamic light scattering (DLS) and atomic force microscopy (AFM) to verify the action and modifications caused in their chemical and physical structures. It was possible to verify the extraction of hemicellulose and lignin, the reduction of fiber sizes to the nanoscale, and the final sample presenting superior crystallinity for the bleached fiber., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
27. A Missense Mutation A384P Associated with Human Hyperekplexia Reveals a Desensitization Site of Glycine Receptors.
- Author
-
Wang CH, Hernandez CC, Wu J, Zhou N, Hsu HY, Shen ML, Wang YC, Macdonald RL, and Wu DC
- Subjects
- Animals, Biotinylation, Cells, Cultured, Excitatory Postsynaptic Potentials genetics, Female, Ganglia, Spinal cytology, Ganglia, Spinal drug effects, Glycine pharmacology, HEK293 Cells, Humans, Male, Patch-Clamp Techniques, Proline genetics, Rats, Rats, Sprague-Dawley, Muscle Rigidity genetics, Mutation, Missense genetics, Receptors, Glycine genetics
- Abstract
Hyperekplexia, an inherited neuronal disorder characterized by exaggerated startle responses with unexpected sensory stimuli, is caused by dysfunction of glycinergic inhibitory transmission. From analysis of newly identified human hyperekplexia mutations in the glycine receptor (GlyR) α1 subunit, we found that an alanine-to-proline missense mutation (A384P) resulted in substantially higher desensitization level and lower agonist sensitivity of homomeric α1 GlyRs when expressed in HEK cells. The incorporation of the β subunit fully reversed the reduction in agonist sensitivity and partially reversed the desensitization of α1
A384P The heteromeric α1A384P β GlyRs showed enhanced desensitization but unchanged agonist-induced maximum responses, surface expression, main channel conductance, and voltage dependence compared with that of the wild-type α1β (α1WT β) GlyRs. Coexpression of the R392H and A384P mutant α1 subunits, which mimic the expression of the compound heterozygous mutation in a hyperekplexia patient, resulted in channel properties similar to those with α1A384P subunit expression alone. In comparison, another human hyperekplexia mutation α1P250T , which was previously reported to enhance desensitization, caused a strong reduction in maximum currents in addition to the altered desensitization. These results were further confirmed by overexpression of α1P250T or α1A384P subunits in cultured neurons isolated from SD rats of either sex. Moreover, the IPSC-like responses of cells expressing α1A384P β induced by repeated glycine pulses showed a stronger frequency-dependent reduction than those expressing α1WT β. Together, our findings demonstrate that A384 is associated with the desensitization site of the α1 subunit and its proline mutation produced enhanced desensitization of GlyRs, which contributes to the pathogenesis of human hyperekplexia. SIGNIFICANCE STATEMENT Human startle disease is caused by impaired synaptic inhibition in the brainstem and spinal cord, which is due to either direct loss of GlyR channel function or reduced number of synaptic GlyRs. Considering that fast decay kinetics of GlyR-mediated inhibitory synaptic responses, the question was raised whether altered desensitization of GlyRs will cause dysfunction of glycine transmission and disease phenotypes. Here, we found that the α1 subunit mutation A384P, identified from startle disease patients, results in enhanced desensitization and leads to rapidly decreasing responses in the mutant GlyRs when they are activated repeatedly by the synaptic-like simulation. These observations suggest that the enhanced desensitization of postsynaptic GlyRs could be the primary pathogenic mechanism of human startle disease., (Copyright © 2018 the authors 0270-6474/18/382819-14$15.00/0.)- Published
- 2018
- Full Text
- View/download PDF
28. GABA A Receptor Coupling Junction and Pore GABRB3 Mutations are Linked to Early-Onset Epileptic Encephalopathy.
- Author
-
Hernandez CC, Zhang Y, Hu N, Shen D, Shen W, Liu X, Kong W, Jiang Y, and Macdonald RL
- Subjects
- Age of Onset, Amino Acid Sequence, Base Sequence, Cell Membrane metabolism, Diazepam pharmacology, HEK293 Cells, Humans, Ion Channel Gating, Protein Subunits chemistry, Protein Subunits genetics, Protein Transport, Receptors, GABA-A chemistry, Epilepsy, Generalized genetics, Mutation genetics, Receptors, GABA-A genetics
- Abstract
GABA
A receptors are brain inhibitory chloride ion channels. Here we show functional analyses and structural simulations for three de novo missense mutations in the GABAA receptor β3 subunit gene (GABRB3) identified in patients with early-onset epileptic encephalopathy (EOEE) and profound developmental delay. We sought to obtain insights into the molecular mechanisms that might link defects in GABAA receptor biophysics and biogenesis to patients with EOEE. The mutant residues are part of conserved structural domains such as the Cys-loop (L170R) and M2-M3 loop (A305V) that form the GABA binding/channel gating coupling junction and the channel pore (T288N), which are functionally coupled during receptor activation. The mutant coupling junction residues caused rearrangements and formation of new hydrogen bonds in the open state, while the mutant pore residue reshaped the pore cavity. Whereas mutant coupling junction residues uncoupled during activation and caused gain of function, the mutant pore residue favoured low conductance receptors and differential sensitivity to diazepam and loss of function. These data reveal novel molecular mechanisms by which EOEE-linked mutations affect GABAA receptor function.- Published
- 2017
- Full Text
- View/download PDF
29. A de novo missense mutation of GABRB2 causes early myoclonic encephalopathy.
- Author
-
Ishii A, Kang JQ, Schornak CC, Hernandez CC, Shen W, Watkins JC, Macdonald RL, and Hirose S
- Subjects
- Brain diagnostic imaging, Brain physiopathology, Crystallography, X-Ray, Electroencephalography, HEK293 Cells, Humans, Infant, Male, Models, Molecular, Mutation, Missense, Opsoclonus-Myoclonus Syndrome physiopathology, Receptors, GABA-A chemistry, Seizures physiopathology, Spasms, Infantile physiopathology, Opsoclonus-Myoclonus Syndrome genetics, Receptors, GABA-A genetics, Seizures genetics, Spasms, Infantile genetics
- Abstract
Background: Early myoclonic encephalopathy (EME), a disease with a devastating prognosis, is characterised by neonatal onset of seizures and massive myoclonus accompanied by a continuous suppression-burst EEG pattern. Three genes are associated with EMEs that have metabolic features. Here, we report a pathogenic mutation of an ion channel as a cause of EME for the first time., Methods: Sequencing was performed for 214 patients with epileptic seizures using a gene panel with 109 genes that are known or suspected to cause epileptic seizures. Functional assessments were demonstrated by using electrophysiological experiments and immunostaining for mutant γ-aminobutyric acid-A (GABA
A ) receptor subunits in HEK293T cells., Results: We discovered a de novo heterozygous missense mutation (c.859A>C [p.Thr287Pro]) in the GABRB2 -encoded β2 subunit of the GABAA receptor in an infant with EME. No GABRB2 mutations were found in three other EME cases or in 166 patients with infantile spasms. GABAA receptors bearing the mutant β2 subunit were poorly trafficked to the cell membrane and prevented γ2 subunits from trafficking to the cell surface. The peak amplitudes of currents from GABAA receptors containing only mutant β2 subunits were smaller than that of those from receptors containing only wild-type β2 subunits. The decrease in peak current amplitude (96.4% reduction) associated with the mutant GABAA receptor was greater than expected, based on the degree to which cell surface expression was reduced (66% reduction)., Conclusion: This mutation has complex functional effects on GABAA receptors, including reduction of cell surface expression and attenuation of channel function, which would significantly perturb GABAergic inhibition in the brain., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.)- Published
- 2017
- Full Text
- View/download PDF
30. Altered Channel Conductance States and Gating of GABA A Receptors by a Pore Mutation Linked to Dravet Syndrome.
- Author
-
Hernandez CC, Kong W, Hu N, Zhang Y, Shen W, Jackson L, Liu X, Jiang Y, and Macdonald RL
- Subjects
- Animals, Cell Membrane metabolism, Cerebral Cortex metabolism, HEK293 Cells, Humans, Inhibitory Postsynaptic Potentials physiology, Mice, Inbred C57BL, Miniature Postsynaptic Potentials physiology, Models, Molecular, Neurons metabolism, Recombinant Proteins genetics, Recombinant Proteins metabolism, Zinc metabolism, Epilepsies, Myoclonic genetics, Mutation, Missense, Receptors, GABA-A genetics, Receptors, GABA-A metabolism
- Abstract
We identified a de novo missense mutation, P302L, in the γ-aminobutyric acid type A (GABA
A ) receptor γ2 subunit gene GABRG2 in a patient with Dravet syndrome using targeted next-generation sequencing. The mutation was in the cytoplasmic portion of the transmembrane segment M2 of the γ2 subunit that faces the pore lumen. GABAA receptor α1 and β3 subunits were coexpressed with wild-type (wt) γ2L or mutant γ2L(P302L) subunits in HEK 293T cells and cultured mouse cortical neurons. We measured currents using whole-cell and single-channel patch clamp techniques, surface and total expression levels using surface biotinylation and Western blotting, and potential structural perturbations in mutant GABAA receptors using structural modeling. The γ2(P302L) subunit mutation produced an ∼90% reduction of whole-cell current by increasing macroscopic desensitization and reducing GABA potency, which resulted in a profound reduction of GABAA receptor-mediated miniature IPSCs (mIPSCs). The conductance of the receptor channel was reduced to 24% of control conductance by shifting the relative contribution of the conductance states from high- to low-conductance levels with only slight changes in receptor surface expression. Structural modeling of the GABAA receptor in the closed, open, and desensitized states showed that the mutation was positioned to slow activation, enhance desensitization, and shift channels to a low-conductance state by reshaping the hour-glass-like pore cavity during transitions between closed, open, and desensitized states. Our study revealed a novel γ2 subunit missense mutation (P302L) that has a novel pathogenic mechanism to cause defects in the conductance and gating of GABAA receptors, which results in hyperexcitability and contributes to the pathogenesis of the genetic epilepsy Dravet syndrome.- Published
- 2017
- Full Text
- View/download PDF
31. De novo GABRG2 mutations associated with epileptic encephalopathies.
- Author
-
Shen D, Hernandez CC, Shen W, Hu N, Poduri A, Shiedley B, Rotenberg A, Datta AN, Leiz S, Patzer S, Boor R, Ramsey K, Goldberg E, Helbig I, Ortiz-Gonzalez XR, Lemke JR, Marsh ED, and Macdonald RL
- Subjects
- Child, Child, Preschool, Electrophysiological Phenomena, Exome, Female, HEK293 Cells, Humans, Male, Mutation, Patch-Clamp Techniques, Phenotype, Drug Resistant Epilepsy genetics, Drug Resistant Epilepsy physiopathology, Epilepsy genetics, Epilepsy physiopathology, Receptors, GABA-A genetics
- Abstract
Epileptic encephalopathies are a devastating group of severe childhood onset epilepsies with medication-resistant seizures and poor developmental outcomes. Many epileptic encephalopathies have a genetic aetiology and are often associated with de novo mutations in genes mediating synaptic transmission, including GABA
A receptor subunit genes. Recently, we performed next generation sequencing on patients with a spectrum of epileptic encephalopathy phenotypes, and we identified five novel (A106T, I107T, P282S, R323W and F343L) and one known (R323Q) de novo GABRG2 pathogenic variants (mutations) in eight patients. To gain insight into the molecular basis for how these mutations contribute to epileptic encephalopathies, we compared the effects of the mutations on the properties of recombinant α1β2γ2L GABAA receptors transiently expressed in HEK293T cells. Using a combination of patch clamp recording, immunoblotting, confocal imaging and structural modelling, we characterized the effects of these GABRG2 mutations on GABAA receptor biogenesis and channel function. Compared with wild-type α1β2γ2L receptors, GABAA receptors containing a mutant γ2 subunit had reduced cell surface expression with altered subunit stoichiometry or decreased GABA-evoked whole-cell current amplitudes, but with different levels of reduction. While a causal role of these mutations cannot be established directly from these results, the functional analysis together with the genetic information suggests that these GABRG2 variants may be major contributors to the epileptic encephalopathy phenotypes. Our study further expands the GABRG2 phenotypic spectrum and supports growing evidence that defects in GABAergic neurotransmission participate in the pathogenesis of genetic epilepsies including epileptic encephalopathies., (© The Author (2016). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)- Published
- 2017
- Full Text
- View/download PDF
32. Correction: Deleterious Rare Variants Reveal Risk for Loss of GABAA Receptor Function in Patients with Genetic Epilepsy and in the General Population.
- Author
-
Hernandez CC, Klassen TL, Jackson LG, Gurba K, Hu N, Noebels JL, and Macdonald RL
- Abstract
[This corrects the article DOI: 10.1371/journal.pone.0162883.].
- Published
- 2016
- Full Text
- View/download PDF
33. Deleterious Rare Variants Reveal Risk for Loss of GABAA Receptor Function in Patients with Genetic Epilepsy and in the General Population.
- Author
-
Hernandez CC, Klassen TL, Jackson LG, Gurba K, Hu N, Noebels JL, and Macdonald RL
- Subjects
- Amino Acid Sequence, Amino Acid Substitution, Case-Control Studies, Genetic Variation, HEK293 Cells, Humans, Ion Channel Gating genetics, Models, Molecular, Mutant Proteins chemistry, Mutation, Missense, Protein Domains, Protein Subunits chemistry, Protein Subunits genetics, Receptors, GABA-A chemistry, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Sequence Homology, Amino Acid, Epilepsy genetics, Epilepsy metabolism, Mutant Proteins genetics, Mutant Proteins metabolism, Mutation, Receptors, GABA-A genetics, Receptors, GABA-A metabolism
- Abstract
Genetic epilepsies (GEs) account for approximately 50% of all seizure disorders, and familial forms include mutations in single GABAA receptor subunit genes (GABRs). In 144 sporadic GE cases (GECs), exome sequencing of 237 ion channel genes identified 520 GABR variants. Among these variants, 33 rare variants in 11 GABR genes were present in 24 GECs. To assess functional risk of variants in GECs, we selected 8 variants found in GABRA, 3 in GABRB, and 3 in GABRG and compared them to 18 variants found in the general population for GABRA1 (n = 9), GABRB3 (n = 7), and GABRG2 (n = 2). To identify deleterious variants and gain insight into structure-function relationships, we studied the gating properties, surface expression and structural perturbations of the 32 variants. Significant reduction of GABAA receptor function was strongly associated with variants scored as deleterious and mapped within the N-terminal and transmembrane domains. In addition, 12 out of 17 variants mapped along the β+/α- GABA binding interface, were associated with reduction in channel gating and were predicted to cause structural rearrangements of the receptor by in silico simulations. Missense or nonsense mutations of GABRA1, GABRB3 and GABRG2 primarily impair subunit biogenesis. In contrast, GABR variants affected receptor function by impairing gating, suggesting that different mechanisms are operating in GABR epilepsy susceptibility variants and disease-causing mutations. The functional impact of single GABR variants found in individuals with sporadic GEs warrants the use of molecular diagnosis and will ultimately improve the treatment of genetic epilepsies by using a personalized approach., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2016
- Full Text
- View/download PDF
34. Reply.
- Author
-
Janve VS, Hernandez CC, Verdier KM, Hu N, and Macdonald RL
- Published
- 2016
- Full Text
- View/download PDF
35. Epileptic encephalopathy de novo GABRB mutations impair γ-aminobutyric acid type A receptor function.
- Author
-
Janve VS, Hernandez CC, Verdier KM, Hu N, and Macdonald RL
- Abstract
Objective: The Epi4K Consortium recently identified 4 de novo mutations in the γ-aminobutyric acid type A (GABA
A ) receptor β3 subunit gene GABRB3 and 1 in the β1 subunit gene GABRB1 in children with one of the epileptic encephalopathies (EEs) Lennox-Gastaut syndrome (LGS) and infantile spasms (IS). Because the etiology of EEs is often unknown, we determined the impact of GABRB mutations on GABAA receptor function and biogenesis., Methods: GABAA receptor α1 and γ2L subunits were coexpressed with wild-type and/or mutant β3 or β1 subunits in HEK 293T cells. Currents were measured using whole cell and single channel patch clamp techniques. Surface and total expression levels were measured using flow cytometry. Potential structural perturbations in mutant GABAA receptors were explored using structural modeling., Results: LGS-associated GABRB3(D120N, E180G, Y302C) mutations located at β+ subunit interfaces reduced whole cell currents by decreasing single channel open probability without loss of surface receptors. In contrast, IS-associated GABRB3(N110D) and GABRB1(F246S) mutations at β- subunit interfaces produced minor changes in whole cell current peak amplitude but altered current deactivation by decreasing or increasing single channel burst duration, respectively. GABRB3(E180G) and GABRB1(F246S) mutations also produced spontaneous channel openings., Interpretation: All 5 de novo GABRB mutations impaired GABAA receptor function by rearranging conserved structural domains, supporting their role in EEs. The primary effect of LGS-associated mutations was reduced GABA-evoked peak current amplitudes, whereas the major impact of IS-associated mutations was on current kinetic properties. Despite lack of association with epilepsy syndromes, our results suggest GABRB1 as a candidate human epilepsy gene. Ann Neurol 2016;79:806-825., (© 2016 American Neurological Association.)- Published
- 2016
- Full Text
- View/download PDF
36. Three epilepsy-associated GABRG2 missense mutations at the γ+/β- interface disrupt GABAA receptor assembly and trafficking by similar mechanisms but to different extents.
- Author
-
Huang X, Hernandez CC, Hu N, and Macdonald RL
- Subjects
- Adenosine Triphosphatases metabolism, Animals, Cells, Cultured, Cerebral Cortex cytology, Computer Simulation, Embryo, Mammalian, Gene Expression Regulation genetics, HEK293 Cells, Humans, Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase pharmacology, Membrane Potentials drug effects, Membrane Potentials genetics, Models, Molecular, Protein Subunits genetics, Protein Transport genetics, Rats, Receptors, GABA-A drug effects, Temperature, Mutation, Missense genetics, Protein Subunits metabolism, Receptors, GABA-A genetics, Receptors, GABA-A metabolism
- Abstract
We compared the effects of three missense mutations in the GABAA receptor γ2 subunit on GABAA receptor assembly, trafficking and function in HEK293T cells cotransfected with α1, β2, and wildtype or mutant γ2 subunits. The mutations R82Q and P83S were identified in families with genetic epilepsy with febrile seizures plus (GEFS+), and N79S was found in a single patient with generalized tonic-clonic seizures (GTCS). Although all three mutations were located in an N-terminal loop that contributes to the γ+/β- subunit-subunit interface, we found that each mutation impaired GABAA receptor assembly to a different extent. The γ2(R82Q) and γ2(P83S) subunits had reduced α1β2γ2 receptor surface expression due to impaired assembly into pentamers, endoplasmic reticulum (ER) retention and degradation. In contrast, γ2(N79S) subunits were efficiently assembled into GABAA receptors with only minimally altered receptor trafficking, suggesting that N79S was a rare or susceptibility variant rather than an epilepsy mutation. Increased structural variability at assembly motifs was predicted by R82Q and P83S, but not N79S, substitution, suggesting that R82Q and P83S substitutions were less tolerated. Membrane proteins with missense mutations that impair folding and assembly often can be "rescued" by decreased temperatures. We coexpressed wildtype or mutant γ2 subunits with α1 and β2 subunits and found increased surface and total levels of both wildtype and mutant γ2 subunits after decreasing the incubation temperature to 30°C for 24h, suggesting that lower temperatures increased GABAA receptor stability. Thus epilepsy-associated mutations N79S, R82Q and P83S disrupted GABAA receptor assembly to different extents, an effect that could be potentially rescued by facilitating protein folding and assembly., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
37. Co-expression of γ2 subunits hinders processing of N-linked glycans attached to the N104 glycosylation sites of GABAA receptor β2 subunits.
- Author
-
Lo WY, Lagrange AH, Hernandez CC, Gurba KN, and Macdonald RL
- Subjects
- Animals, Glycosylation drug effects, HEK293 Cells, Humans, Mice, Mice, Knockout, gamma-Aminobutyric Acid metabolism, gamma-Aminobutyric Acid pharmacology, Gene Expression Regulation, Polysaccharides metabolism, Receptors, GABA biosynthesis, Receptors, GABA-A biosynthesis
- Abstract
GABAA receptors, the major mediators of fast inhibitory neuronal transmission, are heteropentameric glycoproteins assembled from a panel of subunits, usually including α and β subunits with or without a γ2 subunit. The α1β2γ2 receptor is the most abundant GABAA receptor in brain. Co-expression of γ2 with α1 and β2 subunits causes conformational changes, increases GABAA receptor channel conductance, and prolongs channel open times. We reported previously that glycosylation of the three β2 subunit glycosylation sites, N32, N104 and N173, was important for α1β2 receptor channel gating. Here, we examined the hypothesis that steric effects or conformational changes caused by γ2 subunit co-expression alter the glycosylation of partnering β2 subunits. We found that co-expression of γ2 subunits hindered processing of β2 subunit N104 N-glycans in HEK293T cells. This γ2 subunit-dependent effect was strong enough that a decrease of γ2 subunit expression in heterozygous GABRG2 knockout (γ2(+/-)) mice led to appreciable changes in the endoglycosidase H digestion pattern of neuronal β2 subunits. Interestingly, as measured by flow cytometry, γ2 subunit surface levels were decreased by mutating each of the β2 subunit glycosylation sites. The β2 subunit mutation N104Q also decreased GABA potency to evoke macroscopic currents and reduced conductance, mean open time and open probability of single channel currents. Collectively, our data suggested that γ2 subunits interacted with β2 subunit N-glycans and/or subdomains containing the glycosylation sites, and that γ2 subunit co-expression-dependent alterations in the processing of the β2 subunit N104 N-glycans were involved in altering the function of surface GABAA receptors.
- Published
- 2014
- Full Text
- View/download PDF
38. Impaired surface αβγ GABA(A) receptor expression in familial epilepsy due to a GABRG2 frameshift mutation.
- Author
-
Tian M, Mei D, Freri E, Hernandez CC, Granata T, Shen W, Macdonald RL, and Guerrini R
- Subjects
- Adult, Aged, Amino Acid Sequence, Child, Child, Preschool, Electroencephalography, Epilepsies, Myoclonic metabolism, Female, Flow Cytometry, Frameshift Mutation, Genotype, Humans, Immunoblotting, Immunohistochemistry, Male, Microscopy, Confocal, Middle Aged, Molecular Sequence Data, Patch-Clamp Techniques, Pedigree, Phenotype, Receptors, GABA-A biosynthesis, Epilepsies, Myoclonic genetics, Epilepsies, Myoclonic physiopathology, Receptors, GABA-A genetics
- Abstract
The purpose of the study was to explore the pathogenic mechanisms underlying generalized epilepsy and febrile seizures plus (GEFS+) in a family with a novel γ2 subunit gene (GABRG2) frameshift mutation. Four affected and one unaffected individuals carried a c.1329delC GABRG2 mutation resulting in a subunit [γ2S(S443delC)] with a modified and elongated carboxy-terminus that is different from that of the wildtype γ2S subunit. We expressed the wildtype γ2S subunit and the predicted mutant γ2S(S443delC) subunit cDNAs in HEK293T cells and performed immunoblotting, flow cytometry and electrophysiology studies. The mutant subunit was translated as a stable protein that was larger than the wildtype γ2S subunit and was retained in the ER and not expressed on the cell surface membrane, suggesting GABRG2 haploinsufficiency. Peak GABA-evoked currents recorded from cells cotransfected with wildtype α1 and β2 subunits and mutant γ2S subunits were significantly decreased and were comparable to α1β2 receptor currents. S443delC is the first GABR epilepsy mutation predicted to abolish the natural stop codon and produce a stop codon in the 3' UTR that leads to translation of an extended peptide. The GEFS+ phenotype observed in this family is likely caused by γ2S subunit loss-of-function and possibly to dominant-negative suppression of α1β2γ2 receptors. Many GABRG2 truncation mutations result in GEFS+, but the spectrum of phenotypic severity is wider, ranging from asymptomatic individuals to the Dravet syndrome. Mechanisms influencing the severity of the phenotype are therefore complex and difficult to correlate with its demonstrable functional effects., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
39. The GABRG2 nonsense mutation, Q40X, associated with Dravet syndrome activated NMD and generated a truncated subunit that was partially rescued by aminoglycoside-induced stop codon read-through.
- Author
-
Huang X, Tian M, Hernandez CC, Hu N, and Macdonald RL
- Subjects
- Aminoglycosides genetics, Aminoglycosides metabolism, Codon, Terminator, Epilepsies, Myoclonic metabolism, HEK293 Cells, Humans, Protein Subunits metabolism, Receptors, GABA-A metabolism, Codon, Nonsense, Epilepsies, Myoclonic genetics, Nonsense Mediated mRNA Decay, Protein Subunits genetics, Receptors, GABA-A genetics
- Abstract
The GABRG2 nonsense mutation, Q40X, is associated with the severe epilepsy syndrome, Dravet syndrome, and is predicted to generate a premature translation-termination codon (PTC) in the GABA(A) receptor γ2 subunit mRNA in a position that codes for the first amino acid of the mutant subunit. We determined the effects of the mutation on γ2 subunit mRNA and protein synthesis and degradation, as well as on α1β2γ2 GABA(A) receptor assembly, trafficking and surface expression in HEK cells. Using bacterial artificial chromosome (BAC) constructs, we found that γ2(Q40X) subunit mRNA was degraded by nonsense mediated mRNA decay (NMD). Undegraded mutant mRNA was translated to a truncated peptide, likely the signal peptide, which was cleaved further. We also found that mutant γ2(Q40X) subunits did not assemble into functional receptors, thus decreasing GABA-evoked current amplitudes. The GABRG2(Q40X) mutation is one of several epilepsy-associated nonsense mutations that have the potential to be rescued by reading through the PTC, thus restoring full-length protein translation. As a first approach, we investigated the use of the aminoglycoside, gentamicin, to rescue translation of intact mutant subunits by inducing mRNA read-through. In the presence of gentamicin, synthesis of full length γ2 subunits was partially restored, and surface biotinylation and whole cell recording experiments suggested that rescued γ2 subunits could corporate into functional, surface GABA(A) receptors, indicating a possible direction for future therapy., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
40. Pore determinants of KCNQ3 K+ current expression.
- Author
-
Choveau FS, Hernandez CC, Bierbower SM, and Shapiro MS
- Subjects
- Amino Acid Sequence, Amino Acids metabolism, Animals, CHO Cells, Cell Membrane metabolism, Cricetinae, Cricetulus, Electric Conductivity, Hydrophobic and Hydrophilic Interactions, Microscopy, Fluorescence, Molecular Sequence Data, Mutant Proteins chemistry, Mutant Proteins metabolism, Mutation genetics, Protein Multimerization, Protein Stability, Protein Structure, Tertiary, Structural Homology, Protein, Structure-Activity Relationship, Ion Channel Gating, KCNQ3 Potassium Channel chemistry, KCNQ3 Potassium Channel metabolism
- Abstract
KCNQ3 homomeric channels yield very small macroscopic currents compared with other KCNQ channels or KCNQ2/3 heteromers. Two disparate regions of the channels--the C-terminus and the pore region--have been implicated in governing KCNQ current amplitudes. We previously showed that the C-terminus plays a secondary role compared with the pore region. Here, we confirm the critical role of the pore region in determining KCNQ3 currents. We find that mutations at the 312 position in the pore helix of KCNQ3 (I312E, I312K, and I312R) dramatically decreased KCNQ3 homomeric currents as well as heteromeric KCNQ2/3 currents. Evidence that these mutants were expressed in the heteromers includes shifted TEA sensitivity compared with KCNQ2 homomers. To test for differential membrane protein expression, we performed total internal reflection fluorescence imaging, which revealed only small differences that do not underlie the differences in macroscopic currents. To determine whether this mechanism generalizes to other KCNQ channels, we tested the effects of analogous mutations at the conserved I273 position in KCNQ2, with similar results. Finally, we performed homology modeling of the pore region of wild-type and mutant KCNQ3 channels to investigate the putative structural mechanism mediating these results. The modeling suggests that the lack of current in I312E, I312K, and I312R KCNQ3 channels is due to pore helix-selectivity filter interactions that lock the selectivity filter in a nonconductive conformation., (Copyright © 2012 Biophysical Society. Published by Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
41. GABRB3 mutation, G32R, associated with childhood absence epilepsy alters α1β3γ2L γ-aminobutyric acid type A (GABAA) receptor expression and channel gating.
- Author
-
Gurba KN, Hernandez CC, Hu N, and Macdonald RL
- Subjects
- Amino Acid Sequence, Cell Membrane metabolism, Glycosylation, HEK293 Cells, Humans, Membrane Potentials, Molecular Sequence Data, Patch-Clamp Techniques, Protein Binding, Protein Interaction Domains and Motifs, Protein Multimerization, Protein Structure, Quaternary, Protein Structure, Secondary, Receptors, GABA-A chemistry, Sequence Alignment, Synaptic Transmission, Epilepsy, Absence genetics, Mutation, Missense, Receptors, GABA-A genetics, Receptors, GABA-A metabolism, gamma-Aminobutyric Acid physiology
- Abstract
A GABA(A) receptor β3 subunit mutation, G32R, has been associated with childhood absence epilepsy. We evaluated the possibility that this mutation, which is located adjacent to the most N-terminal of three β3 subunit N-glycosylation sites, might reduce GABAergic inhibition by increasing glycosylation of β3 subunits. The mutation had three major effects on GABA(A) receptors. First, coexpression of β3(G32R) subunits with α1 or α3 and γ2L subunits in HEK293T cells reduced surface expression of γ2L subunits and increased surface expression of β3 subunits, suggesting a partial shift from ternary αβ3γ2L receptors to binary αβ3 and homomeric β3 receptors. Second, β3(G32R) subunits were more likely than β3 subunits to be N-glycosylated at Asn-33, but increases in glycosylation were not responsible for changes in subunit surface expression. Rather, both phenomena could be attributed to the presence of a basic residue at position 32. Finally, α1β3(G32R)γ2L receptors had significantly reduced macroscopic current density. This reduction could not be explained fully by changes in subunit expression levels (because γ2L levels decreased only slightly) or glycosylation (because reduction persisted in the absence of glycosylation at Asn-33). Single channel recording revealed that α1β3(G32R)γ2L receptors had impaired gating with shorter mean open time. Homology modeling indicated that the mutation altered salt bridges at subunit interfaces, including regions important for subunit oligomerization. Our results suggest both a mechanism for mutation-induced hyperexcitability and a novel role for the β3 subunit N-terminal α-helix in receptor assembly and gating.
- Published
- 2012
- Full Text
- View/download PDF
42. The GABRA6 mutation, R46W, associated with childhood absence epilepsy, alters 6β22 and 6β2 GABA(A) receptor channel gating and expression.
- Author
-
Hernandez CC, Gurba KN, Hu N, and Macdonald RL
- Subjects
- HEK293 Cells, Humans, Models, Molecular, Protein Structure, Tertiary, Protein Subunits physiology, Receptors, GABA-A physiology, Epilepsy, Absence genetics, Epilepsy, Absence physiopathology, Ion Channel Gating physiology, Mutation, Receptors, GABA-A genetics
- Abstract
A GABA(A) receptor α6 subunit mutation, R46W, was identified as a susceptibility gene that may contribute to the pathogenesis of childhood absence epilepsy (CAE), but the molecular basis for alteration of GABA(A) receptor function is unclear. The R46W mutation is located in a region homologous to a GABA(A) receptor γ2 subunit missense mutation, R82Q, that is associated with CAE and febrile seizures in humans. To determine how this mutation reduces GABAergic inhibition, we expressed wild-type (α6β2γ2L and α6β2δ) and mutant (α6(R46W)β2γ2L and α6(R46W)β2δ) receptors in HEK 293T cells and characterize their whole-cell and single-channel currents, and surface and total levels. We demonstrated that gating and assembly of both α6(R46W)β2γ2L and α6(R46W)β2δ receptors were impaired. Compared to wild-type currents, α6(R46W)β2γ2L and α6(R46W)β2δ receptors had a reduced current density, α6(R46W)β2γ2L currents desensitized to a greater extent and deactivated at a slower rate, α6(R46W)β2δ receptors did not desensitize but deactivated faster and both α6(R46W)β2γ2L and α6(R46W)β2δ single-channel current mean open times and burst durations were reduced. Surface levels of coexpressed α6(R46W), β2 and δ, but not γ2L, subunits were decreased. 'Heterozygous' coexpression of α6(R46W) and α6 subunits with β2 and γ2L subunits produced intermediate macroscopic current amplitudes by increasing incorporation of wild-type and decreasing incorporation of mutant subunits into receptors trafficked to the surface. Finally, these findings suggest that similar to the γ2(R82Q) mutation, the CAE-associated α6(R46W) mutation could cause neuronal disinhibition and thus increase susceptibility to generalized seizures through a reduction of αβγ and αβδ receptor function and expression.
- Published
- 2011
- Full Text
- View/download PDF
43. Glycosylation of {beta}2 subunits regulates GABAA receptor biogenesis and channel gating.
- Author
-
Lo WY, Lagrange AH, Hernandez CC, Harrison R, Dell A, Haslam SM, Sheehan JH, and Macdonald RL
- Subjects
- Cell Line, Endoplasmic Reticulum genetics, Glycosylation, Humans, Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase chemistry, Mutagenesis, Site-Directed, Protein Stability, Protein Structure, Secondary, Protein Subunits genetics, Receptors, GABA-A genetics, Endoplasmic Reticulum metabolism, Ion Channel Gating physiology, Models, Molecular, Protein Subunits metabolism, Receptors, GABA-A metabolism
- Abstract
γ-aminobutyric acid type A (GABA(A)) receptors are heteropentameric glycoproteins. Based on consensus sequences, the GABA(A) receptor β2 subunit contains three potential N-linked glycosylation sites, Asn-32, Asn-104, and Asn-173. Homology modeling indicates that Asn-32 and Asn-104 are located before the α1 helix and in loop L3, respectively, near the top of the subunit-subunit interface on the minus side, and that Asn-173 is located in the Cys-loop near the bottom of the subunit N-terminal domain. Using site-directed mutagenesis, we demonstrated that all predicted β2 subunit glycosylation sites were glycosylated in transfected HEK293T cells. Glycosylation of each site, however, produced specific changes in α1β2 receptor surface expression and function. Although glycosylation of Asn-173 in the Cys-loop was important for stability of β2 subunits when expressed alone, results obtained with flow cytometry, brefeldin A treatment, and endo-β-N-acetylglucosaminidase H digestion suggested that glycosylation of Asn-104 was required for efficient α1β2 receptor assembly and/or stability in the endoplasmic reticulum. Patch clamp recording revealed that mutation of each site to prevent glycosylation decreased peak α1β2 receptor current amplitudes and altered the gating properties of α1β2 receptor channels by reducing mean open time due to a reduction in the proportion of long open states. In addition to functional heterogeneity, endo-β-N-acetylglucosaminidase H digestion and glycomic profiling revealed that surface β2 subunit N-glycans at Asn-173 were high mannose forms that were different from those of Asn-32 and N104. Using a homology model of the pentameric extracellular domain of α1β2 channel, we propose mechanisms for regulation of GABA(A) receptors by glycosylation.
- Published
- 2010
- Full Text
- View/download PDF
44. Ca2+/calmodulin disrupts AKAP79/150 interactions with KCNQ (M-Type) K+ channels.
- Author
-
Bal M, Zhang J, Hernandez CC, Zaika O, and Shapiro MS
- Subjects
- A Kinase Anchor Proteins genetics, Animals, CHO Cells, Cell Membrane metabolism, Cricetinae, Cricetulus, Fluorescence Resonance Energy Transfer, Humans, KCNQ Potassium Channels genetics, Microscopy, Fluorescence, Patch-Clamp Techniques, Phosphatidylinositol 4,5-Diphosphate physiology, Protein Multimerization, Protein Subunits genetics, Protein Subunits metabolism, Transfection, A Kinase Anchor Proteins metabolism, Calcium physiology, Calmodulin physiology, KCNQ Potassium Channels metabolism
- Abstract
M-type channels are localized to neuronal, cardiovascular, and epithelial tissues, where they play critical roles in control of excitability and K(+) transport, and are regulated by numerous receptors via G(q/11)-mediated signals. One pathway shown for KCNQ2 and muscarinic receptors uses PKC, recruited to the channels by A-kinase anchoring protein (AKAP)79/150. As M-type channels can be variously composed of KCNQ1-5 subunits, and M current is known to be regulated by Ca(2+)/calmodulin (CaM) and PIP(2), we probed the generality of AKAP79/150 actions among KCNQ1-5 channels, and the influence of Ca(2+)/CaM and PIP(2) on AKAP79/150 actions. We first examined which KCNQ subunits are targeted by AKAP79 in Chinese hamster ovary (CHO) cells heterologously expressing KCNQ1-5 subunits and AKAP79, using fluorescence resonance energy transfer (FRET) under total internal reflection fluorescence (TIRF) microscopy, and patch-clamp analysis. Donor-dequenching FRET between CFP-tagged KCNQ1-5 and YFP-tagged AKAP79 revealed association of KCNQ2-5, but not KCNQ1, with AKAP79. In parallel with these results, CHO cells stably expressing M(1) receptors studied under perforated patch-clamp showed cotransfection of AKAP79 to "sensitize" KCNQ2/3 heteromers and KCNQ2-5, but not KCNQ1, homomers to muscarinic inhibition, manifested by shifts in the dose-response relations to lower concentrations. The effect on KCNQ4 was abolished by the T553A mutation of the putative PKC phosphorylation site. We then probed the role of CaM and PIP(2) in these AKAP79 actions. TIRF/FRET experiments revealed cotransfection of wild-type, but not dominant-negative (DN), CaM that cannot bind Ca(2+), to disrupt the interaction of YFP-tagged AKAP79(1-153) with CFP-tagged KCNQ2-5. Tonic depletion of PIP(2) by cotransfection of a PIP(2) phosphatase had no effect, and sudden depletion of PIP(2) did not delocalize GFP-tagged AKAP79 from the membrane. Finally, patch-clamp experiments showed cotransfection of wild-type, but not DN, CaM to prevent the AKAP79-mediated sensitization of KCNQ2/3 heteromers to muscarinic inhibition. Thus, AKAP79 acts on KCNQ2-5, but not KCNQ1-containing channels, with effects disrupted by calcified CaM, but not by PIP(2) depletion.
- Published
- 2010
- Full Text
- View/download PDF
45. Modulation of spontaneous and GABA-evoked tonic alpha4beta3delta and alpha4beta3gamma2L GABAA receptor currents by protein kinase A.
- Author
-
Tang X, Hernandez CC, and Macdonald RL
- Subjects
- Action Potentials drug effects, Animals, Biological Clocks drug effects, Cell Line, Humans, Kidney drug effects, Membrane Potentials drug effects, Rats, Action Potentials physiology, Biological Clocks physiology, Cyclic AMP-Dependent Protein Kinases metabolism, Kidney physiology, Membrane Potentials physiology, Receptors, GABA-A metabolism, gamma-Aminobutyric Acid administration & dosage
- Abstract
Protein kinase A (PKA) has been reported to regulate synaptic alphabetagamma gamma-aminobutyric acid type A (GABA(A)) receptor currents, but whether PKA regulates GABA(A) receptor peri- and extrasynaptic tonic currents is unknown. GABA(A) receptors containing alpha4 subunits are important in mediating tonic inhibition and exist as both alpha4betadelta and alpha4betagamma receptors in the brain. To mimic GABA-independent and GABA-dependent tonic currents, we transfected HEK 293T cells with alpha4beta3delta or alpha4beta3gamma2L subunits and recorded spontaneous currents in the absence of applied GABA and steady-state currents in the presence of 1 muM GABA. Both alpha4beta3delta and alpha4beta3gamma2L receptors displayed spontaneous currents, but PKA activation increased spontaneous alpha4beta3delta currents substantially more than spontaneous alpha4beta3gamma2L currents. The increase in spontaneous alpha4beta3delta currents was due to an increase in single-channel open frequency. In contrast, PKA activation did not alter steady-state tonic currents recorded in the presence of 1 muM GABA. We concluded that PKA had a GABA concentration-dependent effect on alpha4beta3delta and alpha4beta3gamma2L currents. In the absence of GABA, spontaneous alpha4beta3delta and, to a lesser extent, alpha4beta3gamma2L currents could provide a basal, tonic current that could be regulated by PKA. With increasing concentrations of extracellular GABA, however, tonic alpha4beta3delta and alpha4beta3gamma2L currents would become more GABA dependent and less PKA sensitive. Thus in brain regions with fluctuating extracellular GABA levels, the dynamic range of GABA-activated tonic currents would be set by PKA and the increase in tonic current produced by increasing GABA would be reduced by PKA-mediated phosphorylation. When ambient GABA reaches micromolar concentrations, PKA would have no effect on steady-state tonic currents.
- Published
- 2010
- Full Text
- View/download PDF
46. Affinity for phosphatidylinositol 4,5-bisphosphate determines muscarinic agonist sensitivity of Kv7 K+ channels.
- Author
-
Hernandez CC, Falkenburger B, and Shapiro MS
- Subjects
- Animals, CHO Cells, Cricetinae, Cricetulus, Humans, KCNQ Potassium Channels metabolism, KCNQ3 Potassium Channel metabolism, Muscarinic Agonists metabolism, KCNQ2 Potassium Channel metabolism, Muscarinic Agonists pharmacology, Phosphatidylinositol 4,5-Diphosphate metabolism, Receptors, Muscarinic metabolism
- Abstract
Kv7 K(+)-channel subunits differ in their apparent affinity for PIP(2) and are differentially expressed in nerve, muscle, and epithelia in accord with their physiological roles in those tissues. To investigate how PIP(2) affinity affects the response to physiological stimuli such as receptor stimulation, we exposed homomeric and heteromeric Kv7.2, 7.3, and 7.4 channels to a range of concentrations of the muscarinic receptor agonist oxotremorine-M (oxo-M) in a heterologous expression system. Activation of M(1) receptors by oxo-M leads to PIP(2) depletion through G(q) and phospholipase C (PLC). Chinese hamster ovary cells were transiently transfected with Kv7 subunits and M(1) receptors and studied under perforated-patch voltage clamp. For Kv7.2/7.3 heteromers, the EC(50) for current suppression was 0.44 +/- 0.08 microM, and the maximal inhibition (Inhib(max)) was 74 +/- 3% (n = 5-7). When tonic PIP(2) abundance was increased by overexpression of PIP 5-kinase, the EC(50) was shifted threefold to the right (1.2 +/- 0.1 microM), but without a significant change in Inhib(max) (73 +/- 4%, n = 5). To investigate the muscarinic sensitivity of Kv7.3 homomers, we used the A315T pore mutant (Kv7.3(T)) that increases whole-cell currents by 30-fold without any change in apparent PIP(2) affinity. Kv7.3(T) currents had a slightly right-shifted EC(50) as compared with Kv7.2/7.3 heteromers (1.0 +/- 0.8 microM) and a strongly reduced Inhib(max) (39 +/- 3%). In contrast, the dose-response curve of homomeric Kv7.4 channels was shifted considerably to the left (66 +/- 8 nM), and Inhib(max) was slightly increased (81 +/- 6%, n = 3-4). We then studied several Kv7.2 mutants with altered apparent affinities for PIP(2) by coexpressing them with Kv7.3(T) subunits to boost current amplitudes. For the lower affinity (Kv7.2 (R463Q)/Kv7.3(T)) or higher affinity (Kv7.2 (R463E)/Kv7.3(T)) channels, the EC(50) and Inhib(max) were similar to Kv7.4 or Kv7.3(T) homomers (0.12 +/- 0.08 microM and 79 +/- 6% [n = 3-4] and 0.58 +/- 0.07 microM and 27 +/- 3% [n = 3-4], respectively). The very low-affinity Kv7.2 (R452E, R459E, and R461E) triple mutant was also coexpressed with Kv7.3(T). The resulting heteromer displayed a very low EC(50) for inhibition (32 +/- 8 nM) and a slightly increased Inhib(max) (83 +/- 3%, n = 3-4). We then constructed a cellular model that incorporates PLC activation by oxo-M, PIP(2) hydrolysis, PIP(2) binding to Kv7-channel subunits, and K(+) current through Kv7 tetramers. We were able to fully reproduce our data and extract a consistent set of PIP(2) affinities.
- Published
- 2009
- Full Text
- View/download PDF
47. Determinants within the turret and pore-loop domains of KCNQ3 K+ channels governing functional activity.
- Author
-
Zaika O, Hernandez CC, Bal M, Tolstykh GP, and Shapiro MS
- Subjects
- Amino Acid Sequence, Animals, Barium pharmacology, Binding Sites, Biotinylation, CHO Cells, Cell Membrane metabolism, Cricetinae, Cricetulus, Electric Conductivity, Gene Expression Regulation, Humans, KCNQ3 Potassium Channel genetics, Microscopy, Fluorescence, Molecular Sequence Data, Mutation, Porosity, Potassium Channel Blockers pharmacology, Protein Stability, Protein Structure, Tertiary, Sequence Homology, Amino Acid, Substrate Specificity, Tetraethylammonium metabolism, KCNQ3 Potassium Channel chemistry, KCNQ3 Potassium Channel metabolism
- Abstract
KCNQ1-5 (Kv7.1-7.5) subunits assemble to form a variety of functional K(+) channels in the nervous system, heart, and epithelia. KCNQ1 and KCNQ4 homomers and KCNQ2/3 heteromers yield large currents, whereas KCNQ2 and KCNQ3 homomers yield small currents. Since the unitary conductance of KCNQ3 is five- to 10-fold greater than that of KCNQ4 or KCNQ1, these differences are even more striking. To test for differential membrane protein expression, we performed biotinylation and total internal reflection fluorescence imaging assays; however, both revealed only small differences among the channels, leading us to investigate other mechanisms at work. We probed the molecular determinants governing macroscopic current amplitudes, with focus on the turret and pore-loop domains of KCNQ1 and KCNQ3. Elimination of the putative N289 glycosylation site in KCNQ1 reduced current density by approximately 56%. A chimera consisting of KCNQ3 with the turret domain (TD) of KCNQ1 increased current density by about threefold. Replacement of the proximal half of the TD in KCNQ3 with that of KCNQ1 increased current density by fivefold. A triple chimera containing the TD of KCNQ1 and the carboxy terminus of KCNQ4 yielded current density 10- or sixfold larger than wild-type KCNQ3 or KCNQ1, respectively, suggesting that the effects on current amplitudes of the TD and the carboxy-terminus are additive. Critical was the role of the intracellular TEA(+)-binding site. The KCNQ3 (A315T) swap increased current density by 10-fold, and the converse KCNQ1 (T311A) swap reduced it by 10-fold. KCNQ3 (A315S) also yielded greatly increased current amplitudes, whereas currents from mutant A315V channels were very small. The KCNQ3 (A315T) mutation increased the sensitivity of the channels to external Ba(2+) block by eight- to 28-fold, consistent with this mutation altering the structure of the selectivity filter. To investigate a structural hypothesis for the effects of these mutations, we performed homology modeling of the pore region of wild-type and mutant KCNQ3 channels, using KvAP as a template. The modeling suggests a critical stabilizing interaction between the pore helix and the selectivity filter that is absent in wild-type KCNQ3 and the A315V mutant, but present in the A315T and A315S mutants. We conclude that KCNQ3 homomers are well expressed at the plasma membrane, but that most wild-type channels are functionally silent, with rearrangements of the pore-loop architecture induced by the presence of a hydroxyl-containing residue at the 315 position "unlocking" the channels into a conductive conformation.
- Published
- 2008
- Full Text
- View/download PDF
48. Homomeric and heteromeric assembly of KCNQ (Kv7) K+ channels assayed by total internal reflection fluorescence/fluorescence resonance energy transfer and patch clamp analysis.
- Author
-
Bal M, Zhang J, Zaika O, Hernandez CC, and Shapiro MS
- Subjects
- Animals, CHO Cells, Cricetinae, Cricetulus, Fluorescence Resonance Energy Transfer methods, Gene Expression, Humans, KCNQ Potassium Channels genetics, Mutation, Patch-Clamp Techniques methods, Protein Structure, Quaternary genetics, Rats, KCNQ Potassium Channels metabolism
- Abstract
M-type K(+) channels, consisting of KCNQ1-5 (Kv7.1-7.5) subunits, form a variety of homomeric and heteromeric channels. Whereas all the subunits can assemble into homomeric channels, the ability of the subunits to assemble into heteromultimers is highly variable. KCNQ3 is widely thought to co-assemble with several other KCNQ subtypes, whereas KCNQ1 and KCNQ2 do not. However, the existence of other subunit assemblies is not well studied. To systematically explore the heteromeric assembly of KCNQ channels in individual living cells, we performed fluorescence resonance energy transfer (FRET) between cyan fluorescent protein- and yellow fluorescent protein-tagged KCNQ subunits expressed in Chinese hamster ovary cells under total internal reflection fluorescence microscopy in which excitation light only penetrates several hundred nanometers into the cell, thus isolating membrane events. We found significant FRET between homomeric subunits as expected from their functional expression in heterologous expression systems. Also as expected from previous work, robust FRET was observed between KCNQ2 and KCNQ3. KCNQ3 and KCNQ4 also showed substantial FRET as did KCNQ4 and KCNQ5. To determine functional assembly of KCNQ4/KCNQ5 heteromers, we performed two types of experiments. In the first, we constructed a mutant tetraethylammonium ion-sensitive KCNQ4 subunit and tested its assembly with KCNQ5 by patch clamp analysis of the tetraethylammonium ion sensitivity of the resulting current; however, those data were not conclusive. In the second, we co-expressed a KCNQ4 (G285S) pore mutant with KCNQ5 and found the former to act as a dominant negative, suggesting co-assembly of the two types of subunits. These data confirm that among the allowed assembly conformations are KCNQ3/4 and KCNQ4/5 heteromers.
- Published
- 2008
- Full Text
- View/download PDF
49. A carboxy-terminal inter-helix linker as the site of phosphatidylinositol 4,5-bisphosphate action on Kv7 (M-type) K+ channels.
- Author
-
Hernandez CC, Zaika O, and Shapiro MS
- Subjects
- Animals, CHO Cells, Cricetinae, Cricetulus, Ion Channel Gating drug effects, KCNQ1 Potassium Channel drug effects, Protein Binding, Ion Channel Gating physiology, KCNQ1 Potassium Channel physiology, Phosphatidylinositol 4,5-Diphosphate pharmacology
- Abstract
The regulation of M-type (KCNQ [Kv7]) K(+) channels by phosphatidylinositol 4,5-bisphosphate (PIP(2)) has perhaps the best correspondence to physiological signaling, but the site of action and structural motif of PIP(2) on these channels have not been established. Using single-channel recordings of chimeras of Kv7.3 and 7.4 channels with highly differential PIP(2) sensitivities, we localized a carboxy-terminal inter-helix linker as the primary site of PIP(2) action. Point mutants within this linker in Kv7.2 and Kv7.3 identified a conserved cluster of basic residues that interact with the lipid using electrostatic and hydrogen bonds. Homology modeling of this putative PIP(2)-binding linker in Kv7.2 and Kv7.3 using the solved structure of Kir2.1 and Kir3.1 channels as templates predicts a structure of Kv7.2 and 7.3 very similar to the Kir channels, and to the seven-beta-sheet barrel motif common to other PIP(2)-binding domains. Phosphoinositide-docking simulations predict affinities and interaction energies in accord with the experimental data, and furthermore indicate that the precise identity of residues in the interacting pocket alter channel-PIP(2) interactions not only by altering electrostatic energies, but also by allosterically shifting the structure of the lipid-binding surface. The results are likely to shed light on the general structural mechanisms of phosphoinositide regulation of ion channels.
- Published
- 2008
- Full Text
- View/download PDF
50. Regulation of neural KCNQ channels: signalling pathways, structural motifs and functional implications.
- Author
-
Hernandez CC, Zaika O, Tolstykh GP, and Shapiro MS
- Subjects
- Amino Acid Motifs physiology, Humans, Phosphatidylinositol 4,5-Diphosphate metabolism, Potassium Channels, Inwardly Rectifying physiology, Superior Cervical Ganglion physiology, KCNQ Potassium Channels chemistry, KCNQ Potassium Channels physiology, Neurons physiology, Signal Transduction physiology
- Abstract
Neural M-type (KCNQ/Kv7) K(+) channels control somatic excitability, bursting and neurotransmitter release throughout the nervous system. Their activity is regulated by multiple signalling pathways. In superior cervical ganglion sympathetic neurons, muscarinic M(1), angiotensin II AT(1), bradykinin B(2) and purinergic P2Y agonists suppress M current (I(M)). Probes of PLC activity show agonists of all four receptors to induce robust PIP(2) hydrolysis. We have grouped these receptors into two related modes of action. One mode involves depletion of phosphatidylinositol 4,5-bisphosphate (PIP(2)) in the membrane, whose interaction with the channels is thought necessary for their function. The other involves IP(3)-mediated intracellular Ca(2+) signals that stimulate PIP(2) synthesis, preventing its depletion, and suppress I(M) via calmodulin. Carbon-fibre amperometry can evaluate the effect of M channel activity on release of neurotransmitter. Consistent with the dominant role of M current in control of neuronal discharge, M channel openers, or blockers, reduced or augmented the evoked release of noradrenaline neurotransmitter from superior cervical ganglion (SCG) neurons, respectively. We seek to localize the subdomains on the channels critical to their regulation by PIP(2). Based on single-channel recordings from chimeras between high-PIP(2) affinity KCNQ3 and low-PIP(2) affinity KCNQ4 channels, we focus on a 57-residue domain within the carboxy-terminus that is a possible PIP(2) binding site. Homology modelling of this domain using the published structure of IRK1 channels as a template predicts a structure very similar to an analogous region in IRK1 channels, and shows a cluster of basic residues in the KCNQ2 domain to correspond to those implicated in PIP(2) regulation of Kir channels. We discuss some important issues dealing with these topics.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.