Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 151
Filtrar
1.
Int J Mol Sci ; 22(13)2021 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-34206649

RESUMEN

Neurons inevitably rely on a proper repertoire and distribution of membrane-bound ion-conducting channels. Among these proteins, the family of hyperpolarization-activated and cyclic nucleotide-gated (HCN) channels possesses unique properties giving rise to the corresponding Ih-current that contributes to various aspects of neural signaling. In mammals, four genes (hcn1-4) encode subunits of HCN channels. These subunits can assemble as hetero- or homotetrameric ion-conducting channels. In order to elaborate on the specific role of the HCN2 subunit in shaping electrical properties of neurons, we applied an Adeno-associated virus (AAV)-mediated, RNAi-based knock-down strategy of hcn2 gene expression both in vitro and in vivo. Electrophysiological measurements showed that HCN2 subunit knock-down resulted in specific yet anticipated changes in Ih-current properties in primary hippocampal neurons and, in addition, corroborated that the HCN2 subunit participates in postsynaptic signal integration. To further address the role of the HCN2 subunit in vivo, we injected recombinant (r)AAVs into the dorsal hippocampus of young adult male mice. Behavioral and biochemical analyses were conducted to assess the contribution of HCN2-containing channels in shaping hippocampal network properties. Surprisingly, knock-down of hcn2 expression resulted in a severe degeneration of the CA1 pyramidal cell layer, which did not occur in mice injected with control rAAV constructs. This finding might pinpoint to a vital and yet unknown contribution of HCN2 channels in establishing or maintaining the proper function of CA1 pyramidal neurons of the dorsal hippocampus.


Asunto(s)
Apoptosis/genética , Región CA1 Hipocampal/metabolismo , Hipocampo/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/deficiencia , Canales de Potasio/deficiencia , Células Piramidales/metabolismo , Factores de Edad , Animales , Región CA1 Hipocampal/patología , Técnicas de Silenciamiento del Gen , Hipocampo/patología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/química , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Inmunohistoquímica , Ratones , Canales de Potasio/química , Canales de Potasio/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Células Piramidales/patología , Interferencia de ARN
2.
Cell Rep ; 36(3): 109404, 2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34289346

RESUMEN

Glutamatergic and GABAergic synaptic transmission controls excitation and inhibition of postsynaptic neurons, whereas activity of ion channels modulates neuronal intrinsic excitability. However, it is unclear how excessive neuronal excitation affects intrinsic inhibition to regain homeostatic stability under physiological or pathophysiological conditions. Here, we report that a seizure-like sustained depolarization can induce short-term inhibition of hippocampal CA3 neurons via a mechanism of membrane shunting. This depolarization-induced shunting inhibition (DShI) mediates a non-synaptic, but neuronal intrinsic, short-term plasticity that is able to suppress action potential generation and postsynaptic responses by activated ionotropic receptors. We demonstrate that the TRESK channel significantly contributes to DShI. Disruption of DShI by genetic knockout of TRESK exacerbates the sensitivity and severity of epileptic seizures of mice, whereas overexpression of TRESK attenuates seizures. In summary, these results uncover a type of homeostatic intrinsic plasticity and its underlying mechanism. TRESK might represent a therapeutic target for antiepileptic drugs.


Asunto(s)
Potenciales de Acción/fisiología , Canales de Potasio/metabolismo , Convulsiones/fisiopatología , Potenciales de Acción/efectos de los fármacos , Animales , Calcio/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Canales Iónicos/metabolismo , Ligandos , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/deficiencia , Canales de Potasio/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Convulsiones/genética , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Ácido gamma-Aminobutírico/metabolismo
3.
Nature ; 591(7850): 431-437, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33505021

RESUMEN

Lysosomes have fundamental physiological roles and have previously been implicated in Parkinson's disease1-5. However, how extracellular growth factors communicate with intracellular organelles to control lysosomal function is not well understood. Here we report a lysosomal K+ channel complex that is activated by growth factors and gated by protein kinase B (AKT) that we term lysoKGF. LysoKGF consists of a pore-forming protein TMEM175 and AKT: TMEM175 is opened by conformational changes in, but not the catalytic activity of, AKT. The minor allele at rs34311866, a common variant in TMEM175, is associated with an increased risk of developing Parkinson's disease and reduces channel currents. Reduction in lysoKGF function predisposes neurons to stress-induced damage and accelerates the accumulation of pathological α-synuclein. By contrast, the minor allele at rs3488217-another common variant of TMEM175, which is associated with a decreased risk of developing Parkinson's disease-produces a gain-of-function in lysoKGF during cell starvation, and enables neuronal resistance to damage. Deficiency in TMEM175 leads to a loss of dopaminergic neurons and impairment in motor function in mice, and a TMEM175 loss-of-function variant is nominally associated with accelerated rates of cognitive and motor decline in humans with Parkinson's disease. Together, our studies uncover a pathway by which extracellular growth factors regulate intracellular organelle function, and establish a targetable mechanism by which common variants of TMEM175 confer risk for Parkinson's disease.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Lisosomas/metabolismo , Complejos Multiproteicos/metabolismo , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Canales de Potasio/metabolismo , Potasio/metabolismo , Animales , Biocatálisis , Neuronas Dopaminérgicas/metabolismo , Femenino , Mutación con Ganancia de Función , Células HEK293 , Humanos , Mutación con Pérdida de Función , Masculino , Ratones , Ratones Noqueados , Destreza Motora , Complejos Multiproteicos/química , Complejos Multiproteicos/deficiencia , Complejos Multiproteicos/genética , Enfermedad de Parkinson/genética , Canales de Potasio/química , Canales de Potasio/deficiencia , Canales de Potasio/genética , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , alfa-Sinucleína/metabolismo
4.
Nat Commun ; 11(1): 4614, 2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32929069

RESUMEN

The suprachiasmatic nucleus (SCN) is a complex structure dependent upon multiple mechanisms to ensure rhythmic electrical activity that varies between day and night, to determine circadian adaptation and behaviours. SCN neurons are exposed to glutamate from multiple sources including from the retino-hypothalamic tract and from astrocytes. However, the mechanism preventing inappropriate post-synaptic glutamatergic effects is unexplored and unknown. Unexpectedly we discovered that TRESK, a calcium regulated two-pore potassium channel, plays a crucial role in this system. We propose that glutamate activates TRESK through NMDA and AMPA mediated calcium influx and calcineurin activation to then oppose further membrane depolarisation and rising intracellular calcium. Hence, in the absence of TRESK, glutamatergic activity is unregulated leading to membrane depolarisation, increased nocturnal SCN firing, inverted basal calcium levels and impaired sensitivity in light induced phase delays. Our data reveals TRESK plays an essential part in SCN regulatory mechanisms and light induced adaptive behaviours.


Asunto(s)
Adaptación Ocular , Oscuridad , Canales de Potasio/metabolismo , Núcleo Supraquiasmático/fisiología , Animales , Conducta Animal , Calcio/metabolismo , Ácido Glutámico/metabolismo , Luz , Potenciales de la Membrana/efectos de la radiación , Ratones Endogámicos C57BL , Canales de Potasio/deficiencia , Transducción de Señal/efectos de la radiación , Núcleo Supraquiasmático/efectos de la radiación
5.
Nat Commun ; 11(1): 480, 2020 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-31980599

RESUMEN

Mutations in the actively expressed, maternal allele of the imprinted KCNK9 gene cause Birk-Barel intellectual disability syndrome (BBIDS). Using a BBIDS mouse model, we identify here a partial rescue of the BBIDS-like behavioral and neuronal phenotypes mediated via residual expression from the paternal Kcnk9 (Kcnk9pat) allele. We further demonstrate that the second-generation HDAC inhibitor CI-994 induces enhanced expression from the paternally silenced Kcnk9 allele and leads to a full rescue of the behavioral phenotype suggesting CI-994 as a promising molecule for BBIDS therapy. Thus, these findings suggest a potential approach to improve cognitive dysfunction in a mouse model of an imprinting disorder.


Asunto(s)
Anomalías Craneofaciales/genética , Anomalías Craneofaciales/metabolismo , Histonas/metabolismo , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Hipotonía Muscular/genética , Hipotonía Muscular/metabolismo , Canales de Potasio/genética , Animales , Conducta Animal , Benzamidas , Encéfalo/metabolismo , Anomalías Craneofaciales/tratamiento farmacológico , Modelos Animales de Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Impresión Genómica , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Discapacidad Intelectual/tratamiento farmacológico , Locus Coeruleus/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Hipotonía Muscular/tratamiento farmacológico , Mutación , Fenotipo , Fenilendiaminas/farmacología , Canales de Potasio/deficiencia , Canales de Potasio/metabolismo , Regulación hacia Arriba/efectos de los fármacos
6.
Exp Anim ; 69(1): 11-17, 2020 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-31292305

RESUMEN

Hyperpolarization-activated cyclic nucleotide-gated potassium channel 1 (HCN1) contribute to spontaneous rhythmic activity in different tissues, including the heart and brain. Deficiency in HCN1 function is associated with sick sinus syndrome in mice and epilepsy in humans. We recently developed Hcn1-deficient rats and found that they exhibit absence epilepsy. While rearing Hcn1-deficient rats, we noticed loose muscle tension and abnormal gait. We therefore evaluated the muscle strength and motor functions of Hcn1-deficient rats. When subjected to the wire hang test, Hcn1-deficient rats fell down more easily than control F344 rats. Grip strength of Hcn1-deficient rats was significantly smaller than F344 rats. In the inclined plane test, they exhibited a smaller maximum angle. In the rotarod test, the latency to fall was shorter for Hcn1-deficient rats than F344 rats. In the footprint analysis, Hcn1-deficient rats exhibited smaller step length and wider step width than F344 rats. Instead of poor motor coordination ability and muscle weakness, Hcn1-deficient rats exhibited normal electromyograms, muscle histology, and deep tendon reflex. These findings suggest that HCN1 channels contribute to motor coordination and muscle strength, and that the muscle weakness of Hcn1-deficient rats results from the involvement not of the peripheral but of the central nervous system.


Asunto(s)
Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/deficiencia , Fuerza Muscular/genética , Debilidad Muscular/genética , Canales de Potasio/deficiencia , Desempeño Psicomotor/fisiología , Animales , Ratas
7.
Sci Transl Med ; 11(519)2019 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-31748231

RESUMEN

The paucity of selective agonists for TWIK-related acid-sensitive K+ 3 (TASK-3) channel, a member of two-pore domain K+ (K2P) channels, has contributed to our limited understanding of its biological functions. By targeting a druggable transmembrane cavity using a structure-based drug design approach, we discovered a biguanide compound, CHET3, as a highly selective allosteric activator for TASK-3-containing K2P channels, including TASK-3 homomers and TASK-3/TASK-1 heteromers. CHET3 displayed potent analgesic effects in vivo in a variety of acute and chronic pain models in rodents that could be abolished pharmacologically or by genetic ablation of TASK-3. We further found that TASK-3-containing channels anatomically define a unique population of small-sized, transient receptor potential cation channel subfamily M member 8 (TRPM8)-, transient receptor potential cation channel subfamily V member 1 (TRPV1)-, or tyrosine hydroxylase (TH)-positive nociceptive sensory neurons and functionally regulate their membrane excitability, supporting CHET3 analgesic effects in thermal hyperalgesia and mechanical allodynia under chronic pain. Overall, our proof-of-concept study reveals TASK-3-containing K2P channels as a druggable target for treating pain.


Asunto(s)
Analgésicos/farmacología , Activación del Canal Iónico , Canales de Potasio/metabolismo , Analgésicos/química , Animales , Biguanidas/química , Biguanidas/farmacología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Activación del Canal Iónico/efectos de los fármacos , Ligandos , Ratones Noqueados , Nocicepción/efectos de los fármacos , Canales de Potasio/deficiencia , Ratas , Reproducibilidad de los Resultados , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Relación Estructura-Actividad
8.
Neurochem Int ; 129: 104486, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31175897

RESUMEN

Neuron function relies on and instructs the development and precise organization of neurovascular units that in turn support circuit activity. However, our understanding of the molecular cues that regulate this relationship remains sparse. Using a high-throughput screening pipeline, we recently identified several new regulators of vascular patterning. Among these was the potassium channel tetramerization domain-containing protein 7 (KCTD7). Mutations in KCTD7 are associated with progressive myoclonic epilepsy, but how KCTD7 regulates neural development and function remains poorly understood. To begin to identify such mechanisms, we focus on mouse retina, a tractable part of the central nervous system that contains precisely ordered neuron subtypes supported by a trilaminar vascular network. We find that deletion of Kctd7 induces defective patterning of the adult retina vascular network, resulting in increased branching, vessel length, and lacunarity. These alterations reflect early and specific defects in vessel development, as emergence of the superficial and deep vascular layers were delayed. These defects are likely due to a role for Kctd7 in inner retina neurons. Kctd7 is absent from vessels but present in neurons in the inner retina, and its deletion resulted in a corresponding increase in the number of bipolar cells in development and increased vessel branching in adults. These alterations were accompanied by retinal function deficits. Together, these data suggest that neuronal Kctd7 drives growth and patterning of the vasculature and that neurovascular interactions may participate in the pathogenesis of KCTD7-related human diseases.


Asunto(s)
Canales de Potasio/fisiología , Vasos Retinianos/fisiología , Animales , Electrorretinografía , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Epilepsias Mioclónicas Progresivas/genética , Canales de Potasio/deficiencia , Canales de Potasio/genética , ARN Mensajero/biosíntesis , Retina/ultraestructura , Células Bipolares de la Retina/patología , Vasos Retinianos/crecimiento & desarrollo , Vasos Retinianos/patología
9.
Front Immunol ; 10: 744, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31024568

RESUMEN

We previously showed that potassium channel tetramerization domain containing 9 (KCTD9) is aberrantly expressed in natural killer (NK) cells in patients with hepatitis B virus-associated acute-on-chronic liver failure and mice with experimental fulminant hepatitis. However, the mechanism underlying the regulation of NK cell function and fulminant hepatitis progression by KCTD9 is unknown. Here, we investigated the role of Kctd9 in regulation of early development, maturation, and function of NK cells using Kctd9-knockout mice. Compared to wild-type mice, Kctd9-deficient mice exhibited impaired NK cell lineage commitment, as evidenced by selective reduction in the refined NK progenitors, and incomplete NK cell maturation, as manifested by a higher proportion of CD11b- NK cells and a lower percentage of CD11b+ NK cells with high proliferative potential. Moreover, Kctd9-depleted NK cells displayed insufficient IFN-γ production, degranulation, and granzyme B production in response to cytokine stimulation, and attenuated cytotoxicity to tumor cells in vitro. The defect in NK cells was further supported by ameliorated liver damage and improved survival in Kctd9-deficient mice following murine hepatitis virus strain-3 (MHV-3) infection, which otherwise leads to immune-mediated fulminant hepatitis, a phenotype homologous to that caused by NK cell depletion in wild-type mice. Further investigation to identify the underlying mechanism revealed that Kctd9 deficiency hindered the expression of transcription factors, including Ets1, Nfil3, Eomes, and Id2 in NK cells. Collectively, our data reveal that Kctd9 acts as a novel regulator for NK cell commitment, maturation, and effector function.


Asunto(s)
Células Asesinas Naturales/metabolismo , Canales de Potasio/deficiencia , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Proliferación Celular , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/patología , Regulación hacia Abajo , Hepatitis Viral Animal/inmunología , Hepatitis Viral Animal/patología , Humanos , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Hígado/inmunología , Hígado/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Virus de la Hepatitis Murina , Canales de Potasio/genética , Canales de Potasio/inmunología , Factores de Transcripción/genética , Factores de Transcripción/inmunología
10.
Acta Neuropathol ; 137(6): 901-918, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30874922

RESUMEN

Emerging evidence suggested a converging mechanism in neurodegenerative brain diseases (NBD) involving early neuronal network dysfunctions and alterations in the homeostasis of neuronal firing as culprits of neurodegeneration. In this study, we used paired-end short-read and direct long-read whole genome sequencing to investigate an unresolved autosomal dominant dementia family significantly linked to 7q36. We identified and validated a chromosomal inversion of ca. 4 Mb, segregating on the disease haplotype and disrupting the coding sequence of dipeptidyl-peptidase 6 gene (DPP6). DPP6 resequencing identified significantly more rare variants-nonsense, frameshift, and missense-in early-onset Alzheimer's disease (EOAD, p value = 0.03, OR = 2.21 95% CI 1.05-4.82) and frontotemporal dementia (FTD, p = 0.006, OR = 2.59, 95% CI 1.28-5.49) patient cohorts. DPP6 is a type II transmembrane protein with a highly structured extracellular domain and is mainly expressed in brain, where it binds to the potassium channel Kv4.2 enhancing its expression, regulating its gating properties and controlling the dendritic excitability of hippocampal neurons. Using in vitro modeling, we showed that the missense variants found in patients destabilize DPP6 and reduce its membrane expression (p < 0.001 and p < 0.0001) leading to a loss of protein. Reduced DPP6 and/or Kv4.2 expression was also detected in brain tissue of missense variant carriers. Loss of DPP6 is known to cause neuronal hyperexcitability and behavioral alterations in Dpp6-KO mice. Taken together, the results of our genomic, genetic, expression and modeling analyses, provided direct evidence supporting the involvement of DPP6 loss in dementia. We propose that loss of function variants have a higher penetrance and disease impact, whereas the missense variants have a variable risk contribution to disease that can vary from high to low penetrance. Our findings of DPP6, as novel gene in dementia, strengthen the involvement of neuronal hyperexcitability and alteration in the homeostasis of neuronal firing as a disease mechanism to further investigate.


Asunto(s)
Inversión Cromosómica , Demencia/genética , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/deficiencia , Mutación , Proteínas del Tejido Nervioso/deficiencia , Enfermedades Neurodegenerativas/genética , Neuronas/fisiología , Canales de Potasio/deficiencia , Potenciales de Acción/fisiología , Adulto , Anciano , Cromosomas Humanos Par 7/genética , Demencia/fisiopatología , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/fisiología , Femenino , Genes Dominantes , Homeostasis , Humanos , Masculino , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Enfermedades Neurodegenerativas/fisiopatología , Linaje , Penetrancia , Polimorfismo de Nucleótido Simple , Canales de Potasio/genética , Canales de Potasio/fisiología , Estabilidad Proteica , Transporte de Proteínas , Transmisión Sináptica , Secuenciación Completa del Genoma
11.
Ann Neurol ; 84(5): 766-780, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30295347

RESUMEN

OBJECTIVE: Several small case series identified KCTD7 mutations in patients with a rare autosomal recessive disorder designated progressive myoclonic epilepsy (EPM3) and neuronal ceroid lipofuscinosis (CLN14). Despite the name KCTD (potassium channel tetramerization domain), KCTD protein family members lack predicted channel domains. We sought to translate insight gained from yeast studies to uncover disease mechanisms associated with deficiencies in KCTD7 of unknown function. METHODS: Novel KCTD7 variants in new and published patients were assessed for disease causality using genetic analyses, cell-based functional assays of patient fibroblasts and knockout yeast, and electron microscopy of patient samples. RESULTS: Patients with KCTD7 mutations can exhibit movement disorders or developmental regression before seizure onset, and are distinguished from similar disorders by an earlier age of onset. Although most published KCTD7 patient variants were excluded from a genome sequence database of normal human variations, most newly identified patient variants are present in this database, potentially challenging disease causality. However, genetic analysis and impaired biochemical interactions with cullin 3 support a causal role for patient KCTD7 variants, suggesting deleterious alleles of KCTD7 and other rare disease variants may be underestimated. Both patient-derived fibroblasts and yeast lacking Whi2 with sequence similarity to KCTD7 have impaired autophagy consistent with brain pathology. INTERPRETATION: Biallelic KCTD7 mutations define a neurodegenerative disorder with lipofuscin and lipid droplet accumulation but without defining features of neuronal ceroid lipofuscinosis or lysosomal storage disorders. KCTD7 deficiency appears to cause an underlying autophagy-lysosome defect conserved in yeast, thereby assigning a biological role for KCTD7. Ann Neurol 2018;84:774-788.


Asunto(s)
Autofagia/genética , Lisosomas/genética , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Canales de Potasio/deficiencia , Edad de Inicio , Preescolar , Femenino , Humanos , Lactante , Lisosomas/patología , Masculino , Mutación , Linaje , Canales de Potasio/genética , Proteínas de Saccharomyces cerevisiae/genética
12.
Nat Neurosci ; 21(2): 270-282, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29335607

RESUMEN

Medial entorhinal cortex (MEC) grid cells fire at regular spatial intervals and project to the hippocampus, where place cells are active in spatially restricted locations. One feature of the grid population is the increase in grid spatial scale along the dorsal-ventral MEC axis. However, the difficulty in perturbing grid scale without impacting the properties of other functionally defined MEC cell types has obscured how grid scale influences hippocampal coding and spatial memory. Here we use a targeted viral approach to knock out HCN1 channels selectively in MEC, causing the grid scale to expand while leaving other MEC spatial and velocity signals intact. Grid scale expansion resulted in place scale expansion in fields located far from environmental boundaries, reduced long-term place field stability and impaired spatial learning. These observations, combined with simulations of a grid-to-place cell model and position decoding of place cells, illuminate how grid scale impacts place coding and spatial memory.


Asunto(s)
Mapeo Encefálico , Corteza Entorrinal/citología , Células de Red/fisiología , Vías Nerviosas/fisiología , Células de Lugar/fisiología , Percepción Espacial/fisiología , Potenciales de Acción/genética , Potenciales de Acción/fisiología , Animales , Animales Recién Nacidos , Electroencefalografía , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/citología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/deficiencia , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Neurológicos , Fosfopiruvato Hidratasa/metabolismo , Canales de Potasio/deficiencia , Canales de Potasio/genética
13.
Endocrinology ; 158(12): 4129-4138, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29069360

RESUMEN

Primary aldosteronism (PA) is a common form of endocrine hypertension that is characterized by the excessive production of aldosterone relative to suppressed plasma renin levels. PA is usually caused by either a unilateral aldosterone-producing adenoma or bilateral adrenal hyperplasia. Somatic mutations have been identified in several genes that encode ion pumps and channels that may explain the aldosterone excess in over half of aldosterone-producing adenomas, whereas the pathophysiology of bilateral adrenal hyperplasia is largely unknown. A number of mouse models of hyperaldosteronism have been described that recreate some features of the human disorder, although none replicate the genetic basis of human PA. Animal models that reproduce the genotype-phenotype associations of human PA are required to establish the functional mechanisms that underlie the endocrine autonomy and deregulated cell growth of the affected adrenal and for preclinical studies of novel therapeutics. Herein, we discuss the differences in adrenal physiology across species and describe the genetically modified mouse models of PA that have been developed to date.


Asunto(s)
Glándulas Suprarrenales/fisiología , Glándulas Suprarrenales/fisiopatología , Modelos Animales de Enfermedad , Hiperaldosteronismo/fisiopatología , Proteína de la Poliposis Adenomatosa del Colon/deficiencia , Proteína de la Poliposis Adenomatosa del Colon/genética , Glándulas Suprarrenales/metabolismo , Animales , Criptocromos/deficiencia , Criptocromos/genética , Humanos , Hiperaldosteronismo/genética , Hiperaldosteronismo/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/deficiencia , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Ratones Noqueados , Ratones Transgénicos , Canales de Potasio/deficiencia , Canales de Potasio/genética , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Canales de Potasio de Dominio Poro en Tándem/genética , Especificidad de la Especie
14.
Am J Physiol Lung Cell Mol Physiol ; 313(6): L1030-L1046, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28839101

RESUMEN

We previously proposed a role for the two-pore domain potassium (K2P) channel TREK-1 in hyperoxia (HO)-induced lung injury. To determine whether redundancy among the three TREK isoforms (TREK-1, TREK-2, and TRAAK) could protect from HO-induced injury, we now examined the effect of deletion of all three TREK isoforms in a clinically relevant scenario of prolonged HO exposure and mechanical ventilation (MV). We exposed WT and TREK-1/TREK-2/TRAAK-deficient [triple knockout (KO)] mice to either room air, 72-h HO, MV [high and low tidal volume (TV)], or a combination of HO + MV and measured quasistatic lung compliance, bronchoalveolar lavage (BAL) protein concentration, histologic lung injury scores (LIS), cellular apoptosis, and cytokine levels. We determined surfactant gene and protein expression and attempted to prevent HO-induced lung injury by prophylactically administering an exogenous surfactant (Curosurf). HO treatment increased lung injury in triple KO but not WT mice, including an elevated LIS, BAL protein concentration, and markers of apoptosis, decreased lung compliance, and a more proinflammatory cytokine phenotype. MV alone had no effect on lung injury markers. Exposure to HO + MV (low TV) further decreased lung compliance in triple KO but not WT mice, and HO + MV (high TV) was lethal for triple KO mice. In triple KO mice, the HO-induced lung injury was associated with decreased surfactant protein (SP) A and SPC but not SPB and SPD expression. However, these changes could not be explained by alterations in the transcription factors nuclear factor-1 (NF-1), NKX2.1/thyroid transcription factor-1 (TTF-1) or c-jun, or lamellar body levels. Prophylactic Curosurf administration did not improve lung injury scores or compliance in triple KO mice.


Asunto(s)
Hiperoxia/metabolismo , Lesión Pulmonar/metabolismo , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Canales de Potasio/deficiencia , Proteínas Asociadas a Surfactante Pulmonar/biosíntesis , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Hiperoxia/genética , Hiperoxia/patología , Lipopolisacáridos/toxicidad , Lesión Pulmonar/genética , Lesión Pulmonar/patología , Ratones , Ratones Noqueados , Proteínas Asociadas a Surfactante Pulmonar/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
Respir Physiol Neurobiol ; 244: 17-25, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28673876

RESUMEN

Despite intensive research, the exact function of TASK potassium channels in central and peripheral chemoreception is still under debate. In this study, we investigated the respiration of unrestrained TASK-3 (TASK-3-/-) and TASK-1/TASK-3 double knockout (TASK-1/3-/-) adult male mice in vivo using a plethysmographic device. Ventilation parameters of TASK-3-/- mice were normal under control condition (21% O2) and upon hypoxia and hypercapnia they displayed the physiological increase of ventilation. TASK-1/3-/- mice showed increased ventilation under control conditions. This increase of ventilation was caused by increased tidal volumes (VT), a phenomenon similarly observed in TASK-1-/- mice. Under acute hypoxia, TASK-1/3-/- mice displayed the physiological increase of the minute volume. Interestingly, this increase was not related to an increase of the respiratory frequency (fR), as observed in wild-type mice, but was caused by a strong increase of VT. This particular respiratory phenotype is reminiscent of the respiratory phenotype of carotid body-denervated rodents in the compensated state. Acute hypercapnia (5% CO2) stimulated ventilation in TASK-1/3-/- and wild-type mice to a similar extent; however, at higher CO2 concentrations (>5% CO2) the stimulation of ventilation was more pronounced in TASK-1/3-/- mice. At hyperoxia (100% O2), TASK-1-/-, TASK-3-/- and wild-type mice showed the physiological small decrease of ventilation. In sharp contrast, TASK-1/3-/- mice exhibited an abnormal increase of ventilation under hyperoxia. In summary, these measurements showed a grossly normal respiration of TASK-3-/- mice and a respiratory phenotype of TASK-1/3-/- mice that was characterized by a markedly enhanced tidal volume, similar to the one observed in TASK-1-/- mice. The abnormal hyperoxia response, exclusively found in TASK-1/3-/- double mutant mice, indicates that both TASK-1 and TASK-3 are essential for the hyperoxia-induced hypoventilation. The peculiar respiratory phenotype of TASK-1/3 knockout mice is reminiscent of the respiration of animals with long-term carotid body dysfunction. Taken together, TASK-1 and TASK-3 appear to serve specific and distinct roles in the complex processes underlying chemoreception and respiratory control.


Asunto(s)
Hiperoxia/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Canales de Potasio/deficiencia , Respiración , Animales , Dióxido de Carbono/metabolismo , Femenino , Hipercapnia/metabolismo , Hipoxia/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Fenotipo , Pletismografía Total , Canales de Potasio/genética , Canales de Potasio de Dominio Poro en Tándem/genética , Volumen de Ventilación Pulmonar/fisiología
16.
Nature ; 546(7657): 307-311, 2017 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-28562590

RESUMEN

As key executers of biological functions, the activity and abundance of proteins are subjected to extensive regulation. Deciphering the genetic architecture underlying this regulation is critical for understanding cellular signalling events and responses to environmental cues. Using random mutagenesis in haploid human cells, we apply a sensitive approach to directly couple genomic mutations to protein measurements in individual cells. Here we use this to examine a suite of cellular processes, such as transcriptional induction, regulation of protein abundance and splicing, signalling cascades (mitogen-activated protein kinase (MAPK), G-protein-coupled receptor (GPCR), protein kinase B (AKT), interferon, and Wingless and Int-related protein (WNT) pathways) and epigenetic modifications (histone crotonylation and methylation). This scalable, sequencing-based procedure elucidates the genetic landscapes that control protein states, identifying genes that cause very narrow phenotypic effects and genes that lead to broad phenotypic consequences. The resulting genetic wiring map identifies the E3-ligase substrate adaptor KCTD5 (ref. 1) as a negative regulator of the AKT pathway, a key signalling cascade frequently deregulated in cancer. KCTD5-deficient cells show elevated levels of phospho-AKT at S473 that could not be attributed to effects on canonical pathway components. To reveal the genetic requirements for this phenotype, we iteratively analysed the regulatory network linked to AKT activity in the knockout background. This genetic modifier screen exposes suppressors of the KCTD5 phenotype and mechanistically demonstrates that KCTD5 acts as an off-switch for GPCR signalling by triggering proteolysis of Gßγ heterodimers dissociated from the Gα subunit. Although biological networks have previously been constructed on the basis of gene expression, protein-protein associations, or genetic interaction profiles, we foresee that the approach described here will enable the generation of a comprehensive genetic wiring map for human cells on the basis of quantitative protein states.


Asunto(s)
Canales de Potasio/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/genética , Análisis de la Célula Individual/métodos , Células Cultivadas , Haploidia , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Histonas/química , Histonas/metabolismo , Humanos , Interferones/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mutagénesis , Fenotipo , Fosforilación/genética , Canales de Potasio/deficiencia , Canales de Potasio/genética , Proteolisis , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Vía de Señalización Wnt
17.
Proc Natl Acad Sci U S A ; 114(9): 2389-2394, 2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-28193887

RESUMEN

Parkinson disease (PD) is a neurodegenerative disorder pathologically characterized by nigrostriatal dopamine neuron loss and the postmortem presence of Lewy bodies, depositions of insoluble α-synuclein, and other proteins that likely contribute to cellular toxicity and death during the disease. Genetic and biochemical studies have implicated impaired lysosomal and mitochondrial function in the pathogenesis of PD. Transmembrane protein 175 (TMEM175), the lysosomal K+ channel, is centered under a major genome-wide association studies peak for PD, making it a potential candidate risk factor for the disease. To address the possibility that variation in TMEM175 could play a role in PD pathogenesis, TMEM175 function was investigated in a neuronal model system. Studies confirmed that TMEM175 deficiency results in unstable lysosomal pH, which led to decreased lysosomal catalytic activity, decreased glucocerebrosidase activity, impaired autophagosome clearance by the lysosome, and decreased mitochondrial respiration. Moreover, TMEM175 deficiency in rat primary neurons resulted in increased susceptibility to exogenous α-synuclein fibrils. Following α-synuclein fibril treatment, neurons deficient in TMEM175 were found to have increased phosphorylated and detergent-insoluble α-synuclein deposits. Taken together, data from these studies suggest that TMEM175 plays a direct and critical role in lysosomal and mitochondrial function and PD pathogenesis and highlight this ion channel as a potential therapeutic target for treating PD.


Asunto(s)
Autofagosomas/metabolismo , Neuronas Dopaminérgicas/metabolismo , Lisosomas/metabolismo , Mitocondrias/metabolismo , Canales de Potasio/genética , alfa-Sinucleína/química , Animales , Autofagosomas/efectos de los fármacos , Autofagosomas/patología , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/patología , Regulación de la Expresión Génica , Glucosilceramidasa/genética , Glucosilceramidasa/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Lisosomas/efectos de los fármacos , Lisosomas/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Modelos Biológicos , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Canales de Potasio/deficiencia , Cultivo Primario de Células , Agregado de Proteínas/efectos de los fármacos , Ratas , alfa-Sinucleína/farmacología
18.
Hear Res ; 345: 57-68, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28065805

RESUMEN

Low-voltage-activated K+ (gKL) and hyperpolarization-activated mixed cation conductances (gh) mediate currents, IKL and Ih, through channels of the Kv1 (KCNA) and HCN families respectively and give auditory neurons the temporal precision required for signaling information about the onset, fine structure, and time of arrival of sounds. Being partially activated at rest, gKL and gh contribute to the resting potential and shape responses to even small subthreshold synaptic currents. Resting gKL and gh also affect the coupling of somatic depolarization with the generation of action potentials. To learn how these important conductances are regulated we have investigated how genetic perturbations affect their expression in octopus cells of the ventral cochlear nucleus (VCN). We report five new findings: First, the magnitude of gh and gKL varied over more than two-fold between wild type strains of mice. Second, average resting potentials are not different in different strains of mice even in the face of large differences in average gKL and gh. Third, IKL has two components, one being α-dendrotoxin (α-DTX)-sensitive and partially inactivating and the other being α-DTX-insensitive, tetraethylammonium (TEA)-sensitive, and non-inactivating. Fourth, the loss of Kv1.1 results in diminution of the α-DTX-sensitive IKL, and compensatory increased expression of an α-DTX-insensitive, tetraethylammonium (TEA)-sensitive IKL. Fifth, Ih and IKL are balanced at the resting potential in all wild type and mutant octopus cells even when resting potentials vary in individual cells over nearly 10 mV, indicating that the resting potential influences the expression of gh and gKL. The independence of resting potentials on gKL and gh shows that gKL and gh do not, over days or weeks, determine the resting potential but rather that the resting potential plays a role in regulating the magnitude of either or both gKL and gh.


Asunto(s)
Vías Auditivas/metabolismo , Núcleo Coclear/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Canal de Potasio Kv.1.1/genética , Potenciales de la Membrana , Canales de Potasio/genética , Animales , Vías Auditivas/citología , Vías Auditivas/efectos de los fármacos , Núcleo Coclear/citología , Núcleo Coclear/efectos de los fármacos , Regulación de la Expresión Génica , Genotipo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/antagonistas & inhibidores , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/deficiencia , Canal de Potasio Kv.1.1/antagonistas & inhibidores , Canal de Potasio Kv.1.1/deficiencia , Potenciales de la Membrana/efectos de los fármacos , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Plasticidad Neuronal , Técnicas de Placa-Clamp , Fenotipo , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/deficiencia , Factores de Tiempo
19.
Behav Brain Res ; 317: 393-400, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27717812

RESUMEN

Gamma-aminobutyric acid (GABA) is the major inhibitory neurotransmitter in the brain and is implicated in the pathophysiology of a number of neuropsychiatric disorders. The GABAB receptors are G-protein coupled receptors consisting of principle subunits and auxiliary potassium channel tetramerization domain (KCTD) subunits. The KCTD subunits 8, 12, 12b and 16 are cytosolic proteins that determine the kinetics of the GABAB receptor response. Previously, we demonstrated that Kctd12 null mutant mice (Kctd12-/-) exhibit increased auditory fear learning and that Kctd12+/- mice show altered circadian activity, as well as increased intrinsic excitability in hippocampal pyramidal neurons. KCTD16 has been demonstrated to influence neuronal excitability by regulating GABAB receptor-mediated gating of postsynaptic ion channels. In the present study we investigated for behavioural endophenotypes in Kctd16-/- and Kctd16+/- mice. Compared with wild-type (WT) littermates, auditory and contextual fear conditioning were normal in both Kctd16-/- and Kctd16+/- mice. When fear memory was tested on the following day, Kctd16-/- mice exhibited less extinction of auditory fear memory relative to WT and Kctd16+/- mice, as well as more contextual fear memory relative to WT and, in particular, Kctd16+/- mice. Relative to WT, both Kctd16+/- and Kctd16-/- mice exhibited normal circadian activity. This study adds to the evidence that auxillary KCTD subunits of GABAB receptors contribute to the regulation of behaviours that could constitute endophenotypes for hyper-reactivity to aversive stimuli in neuropsychiatric disorders.


Asunto(s)
Condicionamiento Clásico/fisiología , Endofenotipos , Memoria/fisiología , Canales de Potasio/deficiencia , Estimulación Acústica , Animales , Ritmo Circadiano/genética , Conducta Exploratoria/fisiología , Miedo/fisiología , Femenino , Genotipo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Canales de Potasio/genética
20.
Exp Anim ; 65(3): 293-301, 2016 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-27026062

RESUMEN

Essential tremor (ET) is a common movement disorder with a poorly understood etiology. The TRM/Kyo mutant rat, showing spontaneous tremor, is an animal model of ET. Recently, we demonstrated that tremors in these rats emerge when two mutant loci, a missense mutation in the hyperpolarization-activated cyclic nucleotide-gated potassium channel 1 (Hcn1) and the tremor (tm) deletion, are present simultaneously. However, we did not identify which gene within the tm deletion causes tremor expression in TRM/Kyo rats. A strong candidate among the 13 genes within the tm deletion is aspartoacylase (Aspa), because some Aspa-knockout mouse strains show tremor. Here, we generated Aspa-knockout rats using transcription activator-like effector nuclease technology and produced Aspa/Hcn1 double-mutant rats by crossing Aspa-knockout rats with Hcn1-mutant rats. The Aspa-knockout rats carried nonsense mutations in exon 4; and ASPA proteins were not detectable in their brain extracts. They showed elevated levels of N-acetyl-L-aspartate (NAA) in urine and spongy vacuolation and abnormal myelination in the central nervous system, but no tremor. By contrast, Aspa/Hcn1 double-mutant rats spontaneously showed tremors resembling those in TRM/Kyo rats, and the tremor was suppressed by drugs therapeutic for ET but not for parkinsonian tremor. These findings indicated that the lack of the Aspa gene caused tremor expression in TRM/Kyo rats. Our animal model suggested that the interaction of NAA accumulation due to ASPA deficiency with an unstable neuronal membrane potential caused by HCN1 deficiency was involved in tremor development.


Asunto(s)
Amidohidrolasas/genética , Codón sin Sentido , Temblor Esencial/genética , Eliminación de Gen , Estudios de Asociación Genética , Mutación Missense , Amidohidrolasas/deficiencia , Animales , Ácido Aspártico/análogos & derivados , Ácido Aspártico/orina , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Epistasis Genética , Temblor Esencial/patología , Exones/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/deficiencia , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Ratones , Canales de Potasio/deficiencia , Canales de Potasio/genética , Ratas , Ratas Mutantes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA