Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 825, 2023 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-36808153

RESUMO

Neuropsychiatric disorders (NPDs) are frequently co-morbid with epilepsy, but the biological basis of shared risk remains poorly understood. The 16p11.2 duplication is a copy number variant that confers risk for diverse NPDs including autism spectrum disorder, schizophrenia, intellectual disability and epilepsy. We used a mouse model of the 16p11.2 duplication (16p11.2dup/+) to uncover molecular and circuit properties associated with this broad phenotypic spectrum, and examined genes within the locus capable of phenotype reversal. Quantitative proteomics revealed alterations to synaptic networks and products of NPD risk genes. We identified an epilepsy-associated subnetwork that was dysregulated in 16p11.2dup/+ mice and altered in brain tissue from individuals with NPDs. Cortical circuits from 16p11.2dup/+ mice exhibited hypersynchronous activity and enhanced network glutamate release, which increased susceptibility to seizures. Using gene co-expression and interactome analysis, we show that PRRT2 is a major hub in the epilepsy subnetwork. Remarkably, correcting Prrt2 copy number rescued aberrant circuit properties, seizure susceptibility and social deficits in 16p11.2dup/+ mice. We show that proteomics and network biology can identify important disease hubs in multigenic disorders, and reveal mechanisms relevant to the complex symptomatology of 16p11.2 duplication carriers.


Assuntos
Transtorno do Espectro Autista , Epilepsia , Deficiência Intelectual , Animais , Camundongos , Transtorno do Espectro Autista/genética , Encéfalo , Deleção Cromossômica , Variações do Número de Cópias de DNA , Epilepsia/genética , Deficiência Intelectual/genética , Proteínas de Membrana/genética , Fenótipo
2.
Biol Psychiatry ; 94(2): 153-163, 2023 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-36581494

RESUMO

BACKGROUND: Schizophrenia (SCZ) is a debilitating psychiatric disorder with a large genetic contribution; however, its neurodevelopmental substrates remain largely unknown. Modeling pathogenic processes in SCZ using human induced pluripotent stem cell-derived neurons (iNs) has emerged as a promising strategy. Copy number variants confer high genetic risk for SCZ, with duplication of the 16p11.2 locus increasing the risk 14.5-fold. METHODS: To dissect the contribution of induced excitatory neurons (iENs) versus GABAergic (gamma-aminobutyric acidergic) neurons (iGNs) to SCZ pathophysiology, we induced iNs from CRISPR (clustered regularly interspaced short palindromic repeats)-Cas9 isogenic and SCZ patient-derived induced pluripotent stem cells and analyzed SCZ-related phenotypes in iEN monocultures and iEN/iGN cocultures. RESULTS: In iEN/iGN cocultures, neuronal firing and synchrony were reduced at later, but not earlier, stages of in vitro development. These were fully recapitulated in iEN monocultures, indicating a primary role for iENs. Moreover, isogenic iENs showed reduced dendrite length and deficits in calcium handling. iENs from 16p11.2 duplication-carrying patients with SCZ displayed overlapping deficits in network synchrony, dendrite outgrowth, and calcium handling. Transcriptomic analysis of both iEN cohorts revealed molecular markers of disease related to the glutamatergic synapse, neuroarchitecture, and calcium regulation. CONCLUSIONS: Our results indicate the presence of 16p11.2 duplication-dependent alterations in SCZ patient-derived iENs. Transcriptomics and cellular phenotyping reveal overlap between isogenic and patient-derived iENs, suggesting a central role of glutamatergic, morphological, and calcium dysregulation in 16p11.2 duplication-mediated pathogenesis. Moreover, excitatory dysfunction during early neurodevelopment is implicated as the basis of SCZ pathogenesis in 16p11.2 duplication carriers. Our results support network synchrony and calcium handling as outcomes directly linked to this genetic risk variant.


Assuntos
Células-Tronco Pluripotentes Induzidas , Esquizofrenia , Humanos , Esquizofrenia/genética , Esquizofrenia/patologia , Cálcio , Neurônios/patologia
3.
Trends Pharmacol Sci ; 43(5): 392-405, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35427475

RESUMO

Induced pluripotent stem cell (iPSC) and gene editing technologies have revolutionized the field of in vitro disease modeling, granting us access to disease-pertinent human cells of the central nervous system. These technologies are particularly well suited for the study of diseases with strong monogenic etiologies. Epilepsy is one of the most common neurological disorders in children, with approximately half of all genetic cases caused by mutations in ion channel genes. These channelopathy-associated epilepsies are clinically diverse, mechanistically complex, and hard to treat. Here, we review the genetic links to epilepsy, the opportunities and challenges of iPSC-based approaches for developing in vitro models of channelopathy-associated disorders, the available tools for effective phenotyping of iPSC-derived neurons, and discuss the potential therapeutic approaches for these devastating diseases.


Assuntos
Canalopatias , Epilepsia , Células-Tronco Pluripotentes Induzidas , Criança , Epilepsia/genética , Epilepsia/terapia , Humanos , Mutação , Neurônios
4.
Stem Cell Reports ; 17(4): 993-1008, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35276091

RESUMO

The ability to precisely edit the genome of human induced pluripotent stem cell (iPSC) lines using CRISPR/Cas9 has enabled the development of cellular models that can address genotype to phenotype relationships. While genome editing is becoming an essential tool in iPSC-based disease modeling studies, there is no established quality control workflow for edited cells. Moreover, large on-target deletions and insertions that occur through DNA repair mechanisms have recently been uncovered in CRISPR/Cas9-edited loci. Yet the frequency of these events in human iPSCs remains unclear, as they can be difficult to detect. We examined 27 iPSC clones generated after targeting 9 loci and found that 33% had acquired large, on-target genomic defects, including insertions and loss of heterozygosity. Critically, all defects had escaped standard PCR and Sanger sequencing analysis. We describe a cost-efficient quality control strategy that successfully identified all edited clones with detrimental on-target events and could facilitate the integrity of iPSC-based studies.


Assuntos
Células-Tronco Pluripotentes Induzidas , Sistemas CRISPR-Cas/genética , Edição de Genes/métodos , Homozigoto , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Controle de Qualidade
5.
Elife ; 102021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33544076

RESUMO

Mutations in KCNQ2, which encodes a pore-forming K+ channel subunit responsible for neuronal M-current, cause neonatal epileptic encephalopathy, a complex disorder presenting with severe early-onset seizures and impaired neurodevelopment. The condition is exceptionally difficult to treat, partially because the effects of KCNQ2 mutations on the development and function of human neurons are unknown. Here, we used induced pluripotent stem cells (iPSCs) and gene editing to establish a disease model and measured the functional properties of differentiated excitatory neurons. We find that patient iPSC-derived neurons exhibit faster action potential repolarization, larger post-burst afterhyperpolarization and a functional enhancement of Ca2+-activated K+ channels. These properties, which can be recapitulated by chronic inhibition of M-current in control neurons, facilitate a burst-suppression firing pattern that is reminiscent of the interictal electroencephalography pattern in patients. Our findings suggest that dyshomeostatic mechanisms compound KCNQ2 loss-of-function leading to alterations in the neurodevelopmental trajectory of patient iPSC-derived neurons.


Assuntos
Encefalopatias/genética , Canal de Potássio KCNQ2/genética , Neurônios/fisiologia , Potenciais de Ação/fisiologia , Encefalopatias/fisiopatologia , Linhagem Celular , Humanos , Canal de Potássio KCNQ2/metabolismo , Células-Tronco Pluripotentes
6.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33593893

RESUMO

Behaviors that rely on the hippocampus are particularly susceptible to chronological aging, with many aged animals (including humans) maintaining cognition at a young adult-like level, but many others the same age showing marked impairments. It is unclear whether the ability to maintain cognition over time is attributable to brain maintenance, sufficient cognitive reserve, compensatory changes in network function, or some combination thereof. While network dysfunction within the hippocampal circuit of aged, learning-impaired animals is well-documented, its neurobiological substrates remain elusive. Here we show that the synaptic architecture of hippocampal regions CA1 and CA3 is maintained in a young adult-like state in aged rats that performed comparably to their young adult counterparts in both trace eyeblink conditioning and Morris water maze learning. In contrast, among learning-impaired, but equally aged rats, we found that a redistribution of synaptic weights amplifies the influence of autoassociational connections among CA3 pyramidal neurons, yet reduces the synaptic input onto these same neurons from the dentate gyrus. Notably, synapses within hippocampal region CA1 showed no group differences regardless of cognitive ability. Taking the data together, we find the imbalanced synaptic weights within hippocampal CA3 provide a substrate that can explain the abnormal firing characteristics of both CA3 and CA1 pyramidal neurons in aged, learning-impaired rats. Furthermore, our work provides some clarity with regard to how some animals cognitively age successfully, while others' lifespans outlast their "mindspans."


Assuntos
Região CA1 Hipocampal/patologia , Região CA3 Hipocampal/patologia , Envelhecimento Cognitivo , Células Piramidais/patologia , Sinapses/patologia , Animais , Masculino , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos F344
7.
Adv Sci (Weinh) ; 6(3): 1801458, 2019 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-30775231

RESUMO

Small interfering ribonucleic acid (siRNA)-based gene knockdown is an effective tool for gene screening and therapeutics. However, the use of nonviral methods has remained an enormous challenge in neural cells. A strategy is reported to design artificial noncationic modular peptides with amplified affinity for siRNA via supramolecular assembly that shows efficient protein knockdown in neural cells. By solid phase synthesis, a sequence that binds specifically double-stranded ribonucleic acid (dsRNA) with a self-assembling peptide for particle formation is integrated. These supramolecular particles can be further functionalized with bioactive sequences without affecting their biophysical properties. The peptide carrier is found to silence efficiently up to 83% of protein expression in primary astroglial and neuronal cell cultures without cytotoxicity. In the case of neurons, a reduction in electrical activity is observed once the presynaptic protein synaptophysin is downregulated by the siRNA-peptide particles. The results demonstrate that the supramolecular particles offer an siRNA delivery platform for efficient nonviral gene screening and discovery of novel therapies for neural cells.

8.
Nano Lett ; 18(10): 6237-6247, 2018 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-30211565

RESUMO

Brain-derived neurotrophic factor (BDNF), a neurotrophin that binds specifically to the tyrosine kinase B (TrkB) receptor, has been shown to promote neuronal differentiation, maturation, and synaptic plasticity in the central nervous system (CNS) during development or after injury and onset of disease. Unfortunately, native BDNF protein-based therapies have had little clinical success due to their suboptimal pharmacological properties. In the past 20 years, BDNF mimetic peptides have been designed with the purpose of activating certain cell pathways that mimic the functional activity of native BDNF, but the interaction of mimetic peptides with cells can be limited due to the conformational specificity required for receptor activation. We report here on the incorporation of a BDNF mimetic sequence into a supramolecular peptide amphiphile filamentous nanostructure capable of activating the BDNF receptor TrkB and downstream signaling in primary cortical neurons in vitro. Interestingly, we found that this BDNF mimetic peptide is only active when displayed on a peptide amphiphile supramolecular nanostructure. We confirmed that increased neuronal maturation is linked to TrkB signaling pathways by analyzing the phosphorylation of downstream signaling effectors and tracking electrical activity over time. Furthermore, three-dimensional gels containing the BDNF peptide amphiphile (PA) nanostructures encourage cell infiltration while increasing functional maturation. Our findings suggest that the BDNF mimetic PA nanostructure creates a highly bioactive matrix that could serve as a biomaterial therapy in injured regions of the CNS. This new strategy has the potential to induce endogenous cell infiltration and promote functional neuronal maturation through the presentation of the BDNF mimetic signal.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Sistema Nervoso Central/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Receptor trkB/genética , Animais , Biomimética , Fator Neurotrófico Derivado do Encéfalo/administração & dosagem , Fator Neurotrófico Derivado do Encéfalo/química , Diferenciação Celular/efeitos dos fármacos , Sistema Nervoso Central/metabolismo , Humanos , Camundongos , Nanoestruturas/administração & dosagem , Nanoestruturas/química , Neurogênese/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/metabolismo , Peptídeos/química , Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Cultura Primária de Células , Transdução de Sinais/efeitos dos fármacos
9.
Epilepsy Curr ; 18(4): 240-245, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30254520

RESUMO

In the current review, we discuss the process of modeling pediatric epileptic encephalopathies with a focus on in vitro iPSC-based technologies. We highlight the potential benefits as well as the challenges of these approaches and propose appropriate standards for the field.

10.
J Bone Miner Res ; 33(10): 1826-1841, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29813186

RESUMO

Andersen's syndrome is a rare disorder affecting muscle, heart, and bone that is associated with mutations leading to a loss of function of the inwardly rectifying K+ channel Kir2.1. Although the Kir2.1 function can be anticipated in excitable cells by controlling the electrical activity, its role in non-excitable cells remains to be investigated. Using Andersen's syndrome-induced pluripotent stem cells, we investigated the cellular and molecular events during the osteoblastic and chondrogenic differentiation that are affected by the loss of the Ik1 current. We show that loss of Kir2.1 channel function impairs both osteoblastic and chondrogenic processes through the downregulation of master gene expression. This downregulation is the result of an impairment of the bone morphogenetic proteins signaling pathway through dephosphorylation of the Smad proteins. Restoring Kir2.1 channel function in Andersen's syndrome cells rescued master genes expression and restored normal osteoblast and chondrocyte behavior. Our results show that Kir2.1-mediated activity controls endochondral and intramembranous ossification signaling pathways. © 2018 American Society for Bone and Mineral Research.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Condrogênese/genética , Regulação da Expressão Gênica , Osteogênese/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Transdução de Sinais/genética , Síndrome de Andersen/genética , Síndrome de Andersen/patologia , Biomarcadores/metabolismo , Diferenciação Celular , Condrócitos/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Matriz Extracelular/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Modelos Biológicos , Osteoblastos/metabolismo , Fosforilação , Proteína Smad1/metabolismo
11.
Cell Tissue Res ; 371(2): 309-323, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29018970

RESUMO

Andersen's syndrome (AS) is a rare autosomal disorder that has been defined by the triad of periodic paralysis, cardiac arrhythmia, and developmental anomalies. AS has been directly linked to over 40 different autosomal dominant negative loss-of-function mutations in the KCNJ2 gene, encoding for the tetrameric strong inward rectifying K+ channel KIR2.1. While KIR2.1 channels have been suggested to contribute to setting the resting membrane potential (RMP) and to control the duration of the action potential (AP) in skeletal and cardiac muscle, the mechanism by which AS mutations produce such complex pathophysiological symptoms is poorly understood. Thus, we use an adenoviral transduction strategy to study in vivo subcellular distribution of wild-type (WT) and AS-associated mutant KIR2.1 channels in mouse skeletal muscle. We determined that WT and D71V AS mutant KIR2.1 channels are localized to the sarcolemma and the transverse tubules (T-tubules) of skeletal muscle fibers, while the ∆314-315 AS KIR2.1 mutation prevents proper trafficking of the homo- or hetero-meric channel complexes. Whole-cell voltage-clamp recordings in individual skeletal muscle fibers confirmed the reduction of inwardly rectifying K+ current (IK1) after transduction with ∆314-315 KIR2.1 as compared to WT channels. Analysis of skeletal muscle function revealed reduced force generation during isometric contraction as well as reduced resistance to muscle fatigue in extensor digitorum longus muscles transduced with AS mutant KIR2.1. Together, these results suggest that KIR2.1 channels may be involved in the excitation-contraction coupling process required for proper skeletal muscle function. Our findings provide clues to mechanisms associated with periodic paralysis in AS.


Assuntos
Síndrome de Andersen/genética , Técnicas de Silenciamento de Genes , Músculo Esquelético/patologia , Mutação/genética , Canais de Potássio Corretores do Fluxo de Internalização/genética , Adenoviridae/metabolismo , Síndrome de Andersen/patologia , Síndrome de Andersen/fisiopatologia , Animais , Células COS , Chlorocebus aethiops , Proteínas de Fluorescência Verde/metabolismo , Humanos , Ativação do Canal Iônico , Contração Isométrica , Camundongos , Fadiga Muscular , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/fisiopatologia
12.
Front Syst Neurosci ; 10: 52, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27375440

RESUMO

Aging-related cognitive deficits have been attributed to dysfunction of neurons due to failures at synaptic or intrinsic loci, or both. Given the importance of the hippocampus for successful encoding of memory and that the main output of the hippocampus is via the CA1 pyramidal neurons, much of the research has been focused on identifying the aging-related changes of these CA1 pyramidal neurons. We and others have discovered that the postburst afterhyperpolarization (AHP) following a train of action potentials is greatly enlarged in CA1 pyramidal neurons of aged animals. This enlarged postburst AHP is a significant factor in reducing the intrinsic excitability of these neurons, and thus limiting their activity in the neural network during learning. Based on these data, it has largely been thought that aging-related cognitive deficits are attributable to reduced activity of pyramidal neurons. However, recent in vivo and ex vivo studies provide compelling evidence that aging-related deficits could also be due to a converse change in CA3 pyramidal neurons, which show increased activity with aging. In this review, we will incorporate these recent findings and posit that an interdependent dynamic dysfunctional change occurs within the hippocampal network, largely due to altered intrinsic excitability in CA1 and CA3 hippocampal pyramidal neurons, which ultimately leads to the aging-related cognitive deficits.

13.
J Neurosci ; 35(38): 13206-18, 2015 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-26400949

RESUMO

Aging-related impairments in hippocampus-dependent cognition have been attributed to maladaptive changes in the functional properties of pyramidal neurons within the hippocampal subregions. Much evidence has come from work on CA1 pyramidal neurons, with CA3 pyramidal neurons receiving comparatively less attention despite its age-related hyperactivation being postulated to interfere with spatial processing in the hippocampal circuit. Here, we use whole-cell current-clamp to demonstrate that aged rat (29-32 months) CA3 pyramidal neurons fire significantly more action potentials (APs) during theta-burst frequency stimulation and that this is associated with faster AP repolarization (i.e., narrower AP half-widths and enlarged fast afterhyperpolarization). Using a combination of patch-clamp physiology, pharmacology, Western blot analyses, immunohistochemistry, and array tomography, we demonstrate that these faster AP kinetics are mediated by enhanced function and expression of Kv4.2/Kv4.3 A-type K(+) channels, particularly within the perisomatic compartment, of CA3 pyramidal neurons. Thus, our study indicates that inhibition of these A-type K(+) channels can restore the intrinsic excitability properties of aged CA3 pyramidal neurons to a young-like state. Significance statement: Age-related learning deficits have been attributed, in part, to altered hippocampal pyramidal neuronal function with normal aging. Much evidence has come from work on CA1 neurons, with CA3 neurons receiving comparatively less attention despite its age-related hyperactivation being postulated to interfere with spatial processing. Hence, we conducted a series of experiments to identify the cellular mechanisms that underlie the hyperexcitability reported in the CA3 region. Contrary to CA1 neurons, we demonstrate that postburst afterhyperpolarization is not altered with aging and that aged CA3 pyramidal neurons are able to fire significantly more action potentials and that this is associated with faster action potential repolarization through enhanced expression of Kv4.2/Kv4.3 A-type K(+) channels, particularly within the cell bodies of CA3 pyramidal neurons.


Assuntos
Envelhecimento/fisiologia , Região CA3 Hipocampal/citologia , Potenciais da Membrana/fisiologia , Células Piramidais/fisiologia , Canais de Potássio Shal/metabolismo , Análise de Variância , Animais , Biofísica , Relação Dose-Resposta a Droga , Estimulação Elétrica , Técnicas In Vitro , Masculino , Potenciais da Membrana/efeitos dos fármacos , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Células Piramidais/efeitos dos fármacos , Ratos , Ratos Endogâmicos F344 , Potenciais Sinápticos/efeitos dos fármacos , Potenciais Sinápticos/fisiologia
14.
Front Pharmacol ; 2: 63, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22016737

RESUMO

Five inherited human disorders affecting skeletal muscle contraction have been traced to mutations in the gene encoding the voltage-gated sodium channel Na(v)1.4. The main symptoms of these disorders are myotonia or periodic paralysis caused by changes in skeletal muscle fiber excitability. Symptoms of these disorders vary from mild or latent disease to incapacitating or even death in severe cases. As new human sodium channel mutations corresponding to disease states become discovered, the importance of understanding the role of the sodium channel in skeletal muscle function and disease state grows.

15.
J Physiol ; 589(Pt 13): 3115-24, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21521764

RESUMO

Myotonia is an intrinsic muscular disorder caused by muscle fibre hyperexcitability, which produces a prolonged time for relaxation after voluntary muscle contraction or internal mechanical stimulation. Missense mutations in skeletal muscle genes encoding Cl− or Na+ channels cause non-dystrophic myotonias.Mutations of the SCN4A gene that encodes the skeletal voltage-gated Na+ channel Nav1.4 can produce opposing phenotypes leading to hyperexcitable or inexcitable muscle fibres. Nav1.4 mutations result in different forms of myotonias that can be found in adults. However, the recently reported myotonic manifestations in infants have been shown to be lethal. This was typically the case for children suffering from severe neonatal episodic laryngospasm (SNEL). A novel Nav1.4 channel missense mutation was found in these children that has not yet been analysed. In this study, we characterize the functional consequences of the new A799S Na+ channel mutation that is associated with sodium channel myotonia in newborn babies. We have used mammalian cell expression and patch-clamp techniques to monitor the channel properties.We found that the A799S substitution changes several biophysical properties of the channel by causing a hyperpolarizing shift of the steady-state activation, and slowing the kinetics of fast inactivation and deactivation. In addition, the single channel open probability was dramatically increased, contributing hence to a severe phenotype. We showed that substitutions at position 799 of the Nav1.4 channel favoured the channel open state with sustained activity leading to hyperexcitability of laryngeal muscles that could be lethal during infancy.


Assuntos
Músculo Esquelético/fisiologia , Mutação de Sentido Incorreto/genética , Canais de Sódio/genética , Substituição de Aminoácidos/genética , Animais , Linhagem Celular , Humanos , Laringismo/genética , Músculo Esquelético/patologia , Miotonia/genética , Canal de Sódio Disparado por Voltagem NAV1.4 , Índice de Gravidade de Doença , Canais de Sódio/efeitos adversos
16.
J Physiol ; 586(23): 5651-63, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18845607

RESUMO

TREK-2 expressed in mammalian cells exhibits small ( approximately 52 pS) and large ( approximately 220 pS) unitary conductance levels. Here we tested the role of the N-terminus (69 amino acids long) in the control of the unitary conductance, and role of the alternative translation initiation as a mechanism that produces isoforms of TREK-2 that show different conductance levels. Deletion of the first half (Delta1-36) of the N-terminus had no effect. However, deletion of most of the N-terminus (Delta1-66) resulted in the appearance of only the large-conductance channel ( approximately 220 pS). In support of the critical function of the distal half of the N-terminus, the deletion mutants Delta1-44 and Delta1-54 produced approximately 90 pS and 188 pS channels, respectively. In Western blot analysis, TREK-2 antibody detected two immunoreactive bands at approximately 54 kDa and approximately 60 kDa from cells expressing wild-type TREK-2 that has three potential translation initiation sites (designated M(1)M(2)M(3)) within the N-terminus. Mutation of the second and third initiation sites from Met to Leu (M(1)L(2)L(3)) produced only the approximately 60 kDa isoform and the small-conductance channel ( approximately 52 pS). Mutants designed to produce translation from the second (M(2)L(3)) or third (M(3)) initiation site produced the approximately 54 kDa isoform, and the large conductance channel ( approximately 185-224 pS). M(1)L(2)L(3), M(2)L(3) and M(3) were relatively selectively permeable to K(+), as judged by the 51-55 mV shifts in reversal potential following a 10-fold change in [K(+)](o). P(Na)/P(K) values were also similar for M(1)L(2)L(3) ( approximately 0.02), M(2)L(3) ( approximately 0.02) and M(3) ( approximately 0.03). Arachidonic acid, proton and membrane stretch activated, whereas dibutyryl-cAMP inhibited all three isoforms of TREK-2, indicating that deletion of the N-terminus does not abolish modulation. These results show that the small and large conductance TREK-2 channels are produced as a result of alternative translation initiation, producing isoforms with long and short N-termini, and that the distal half of the N-terminus controls the unitary conductance.


Assuntos
Ativação do Canal Iônico/fisiologia , Iniciação Traducional da Cadeia Peptídica , Canais de Potássio de Domínios Poros em Tandem/fisiologia , Animais , Ácido Araquidônico/farmacologia , Células COS , Chlorocebus aethiops , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Potenciais da Membrana/fisiologia , Mutação , Fosforilação/efeitos dos fármacos , Potássio/metabolismo , Canais de Potássio/genética , Canais de Potássio/metabolismo , Canais de Potássio/fisiologia , Canais de Potássio de Domínios Poros em Tandem/genética , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Pressão , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiologia , Ratos , Sódio/metabolismo , Transfecção
17.
Am J Physiol Cell Physiol ; 295(1): C92-9, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18495815

RESUMO

Membrane phosphatidylinositol-4,5-bisphosphate (PIP2) is critical for the function of many transient receptor potential (TRP) ion channels. The role of PIP2 in TRPA1 function is not well known. The effect of PIP2 on TRPA1 was investigated by direct application of PIP2 and by using polylysine and PIP2 antibody that sequester PIP2. In inside-out patches from HeLa cells expressing mouse TRPA1, polytriphosphate (PPPi) was added to the bath solution to keep TRPA1 sensitive to allyl isothiocyanate (AITC; mustard oil). Direct application of PIP2 (10 microM) to inside-out patches did not activate TRPA1, but AITC and Delta(9)-tetrahydrocannabinol (THC) produced strong activation. In inside-out patches in which TRPA1 was first activated with AITC (in the presence of PPPi), further addition of PIP2 produced a concentration-dependent inhibition of TRPA1 [agonist concentration producing half-maximal activity (K(1/2)), 2.8 microM]. Consistent with the inhibition of TRPA1 by PIP2, AITC activated a large whole cell current when polylysine or PIP2 antibody was added to the pipette but a markedly diminished current when PIP2 was added to the pipette. In inside-out patches with PPPi in the bath solution, application of PIP2 antibody or polylysine caused activation of TRPA1, and this was blocked by PIP2. However, TRPA1 was not activated by polylysine and PIP2 antibody under whole cell conditions, suggesting a more complex regulation of TRPA1 by PIP2 in intact cells. These results show that PIP2 inhibits TRPA1 and reduces the sensitivity of TRPA1 to AITC.


Assuntos
Fosfatidilinositol 4,5-Difosfato/fisiologia , Canais de Potencial de Receptor Transitório/antagonistas & inibidores , Compostos Alílicos/farmacologia , Animais , Anticorpos/farmacologia , Dronabinol/farmacologia , Células HeLa , Humanos , Ativação do Canal Iônico , Isocianatos/farmacologia , Camundongos , Técnicas de Patch-Clamp , Fosfatidilinositol 4,5-Difosfato/imunologia , Fosfatidilinositol 4,5-Difosfato/farmacologia , Polilisina/farmacologia , Polifosfatos/farmacologia , Canal de Cátion TRPA1 , Canais de Potencial de Receptor Transitório/fisiologia
18.
Brain Res ; 1083(1): 14-20, 2006 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-16542642

RESUMO

Epilepsy is an often-debilitating disease with many etiologies. Genetic predisposition is common for many of the generalized epilepsy syndromes, and mutations in genes encoding neuronal ion channels are causative in many cases. We previously identified a locus for juvenile audiogenic monogenic seizures (jams1) in the Black Swiss mouse strain, delimited by the gene basigin (Bsg) and the marker D10Mit140. This region includes Hcn2, the gene encoding the hyperpolarization-activated cyclic nucleotide-gated channel subunit 2 (HCN2), an ion channel implicated in epilepsy. By sequencing genomic DNA, we found that Black Swiss mice have a single polymorphism in exon 2 within the Hcn2 gene. This single G/C to A/T base change alters the third position of a codon specifying alanine residue 293, without changing the predicted amino acid sequence. Furthermore, we found no detectable differences in HCN2 protein expression in the brains of Black Swiss mice, compared to control mice. We therefore reason that juvenile audiogenic seizures in Black Swiss mice are unlikely to be due to abnormalities of HCN2 channel function.


Assuntos
Encéfalo/metabolismo , Epilepsia Reflexa/genética , Predisposição Genética para Doença/genética , Canais Iônicos/genética , Fatores Etários , Envelhecimento/genética , Sequência de Aminoácidos/genética , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiopatologia , Células COS , Chlorocebus aethiops , Modelos Animais de Doenças , Epilepsia Reflexa/metabolismo , Epilepsia Reflexa/fisiopatologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Canais Iônicos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mutação Puntual/genética , Canais de Potássio , Transmissão Sináptica/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...