Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34903668

RESUMEN

Fast oscillations in cortical circuits critically depend on GABAergic interneurons. Which interneuron types and populations can drive different cortical rhythms, however, remains unresolved and may depend on brain state. Here, we measured the sensitivity of different GABAergic interneurons in prefrontal cortex under conditions mimicking distinct brain states. While fast-spiking neurons always exhibited a wide bandwidth of around 400 Hz, the response properties of spike-frequency adapting interneurons switched with the background input's statistics. Slowly fluctuating background activity, as typical for sleep or quiet wakefulness, dramatically boosted the neurons' sensitivity to gamma and ripple frequencies. We developed a time-resolved dynamic gain analysis and revealed rapid sensitivity modulations that enable neurons to periodically boost gamma oscillations and ripples during specific phases of ongoing low-frequency oscillations. This mechanism predicts these prefrontal interneurons to be exquisitely sensitive to high-frequency ripples, especially during brain states characterized by slow rhythms, and to contribute substantially to theta-gamma cross-frequency coupling.


Asunto(s)
Ritmo Gamma/fisiología , Interneuronas/fisiología , Corteza Prefrontal/citología , Ritmo Teta/fisiología , Animales , Femenino , Masculino , Ratones , Red Nerviosa/fisiología , Técnicas de Placa-Clamp
2.
J Cell Physiol ; 232(8): 2019-2032, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27255432

RESUMEN

Several reports credit mibefradil with tumor suppressing properties arising from its known inhibition of Ca2+ currents. Given that mibefradil (Mb) is also known to inhibit K+ channels, we decided to study the interaction between this organic compound and the tumor-related Kv10.1 channel. Here we report that Mb modulates the gating of Kv10.1. Mb induces an apparent inactivation from both open and early closed states where the channels dwell at hyperpolarized potentials. Additionally, Mb accelerates the kinetics of current activation, in a manner that depends on initial conditions. Our observations suggest that Mb binds to the voltage sensor domain of Kv10.1 channels, thereby modifying the gating of the channels in a way that in some, but not all, aspects opposes to the gating effects exerted by divalent cations. J. Cell. Physiol. 232: 2019-2032, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Activación del Canal Iónico/efectos de los fármacos , Mibefradil/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Sitios de Unión , Canales de Potasio Éter-A-Go-Go/genética , Canales de Potasio Éter-A-Go-Go/metabolismo , Células HEK293 , Humanos , Cinética , Potenciales de la Membrana , Mibefradil/metabolismo , Modelos Biológicos , Bloqueadores de los Canales de Potasio/metabolismo , Unión Proteica , Transfección
3.
Circ Res ; 117(5): 401-12, 2015 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-26078285

RESUMEN

RATIONALE: Monitoring and controlling cardiac myocyte activity with optogenetic tools offer exciting possibilities for fundamental and translational cardiovascular research. Genetically encoded voltage indicators may be particularly attractive for minimal invasive and repeated assessments of cardiac excitation from the cellular to the whole heart level. OBJECTIVE: To test the hypothesis that cardiac myocyte-targeted voltage-sensitive fluorescence protein 2.3 (VSFP2.3) can be exploited as optogenetic tool for the monitoring of electric activity in isolated cardiac myocytes and the whole heart as well as function and maturity in induced pluripotent stem cell-derived cardiac myocytes. METHODS AND RESULTS: We first generated mice with cardiac myocyte-restricted expression of VSFP2.3 and demonstrated distinct localization of VSFP2.3 at the t-tubulus/junctional sarcoplasmic reticulum microdomain without any signs for associated pathologies (assessed by echocardiography, RNA-sequencing, and patch clamping). Optically recorded VSFP2.3 signals correlated well with membrane voltage measured simultaneously by patch clamping. The use of VSFP2.3 for human action potential recordings was confirmed by simulation of immature and mature action potentials in murine VSFP2.3 cardiac myocytes. Optical cardiograms could be monitored in whole hearts ex vivo and minimally invasively in vivo via fiber optics at physiological heart rate (10 Hz) and under pacing-induced arrhythmia. Finally, we reprogrammed tail-tip fibroblasts from transgenic mice and used the VSFP2.3 sensor for benchmarking functional and structural maturation in induced pluripotent stem cell-derived cardiac myocytes. CONCLUSIONS: We introduce a novel transgenic voltage-sensor model as a new method in cardiovascular research and provide proof of concept for its use in optogenetic sensing of physiological and pathological excitation in mature and immature cardiac myocytes in vitro and in vivo.


Asunto(s)
Potenciales de la Membrana/fisiología , Miocitos Cardíacos/fisiología , Optogenética/métodos , Animales , Humanos , Ratones , Ratones Transgénicos , Imagen de Colorante Sensible al Voltaje/métodos
4.
Eur Biophys J ; 45(7): 721-733, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27444284

RESUMEN

The Kv10.1 (Eag1) voltage-gated potassium channel represents a promising molecular target for novel cancer therapies or diagnostic purposes. Physiologically, it is only expressed in the brain, but it was found overexpressed in more than 70 % of tumours of diverse origin. Furthermore, as a plasma membrane protein, it is easily accessible to extracellular interventions. In this study we analysed the feasibility of the anti-Kv10.1 monoclonal antibody mAb62 to target tumour cells in vitro and in vivo and to deliver therapeutics to the tumour. Using time-domain near infrared fluorescence (NIRF) imaging in a subcutaneous MDA-MB-435S tumour model in nude mice, we showed that mAb62-Cy5.5 specifically accumulates at the tumour for at least 1 week in vivo with a maximum intensity at 48 h. Blocking experiments with an excess of unlabelled mAb62 and application of the free Cy5.5 fluorophore demonstrate specific binding to the tumour. Ex vivo NIRF imaging of whole tumours as well as NIRF imaging and microscopy of tumour slices confirmed the accumulation of the mAb62-Cy5.5 in tumours but not in brain tissue. Moreover, mAb62 was conjugated to the prodrug-activating enzyme ß-D-galactosidase (ß-gal; mAb62-ß-gal). The ß-gal activity of the mAb62-ß-gal conjugate was analysed in vitro on Kv10.1-expressing MDA-MB-435S cells in comparison to control AsPC-1 cells. We show that the mAb62-ß-gal conjugate possesses high ß-gal activity when bound to Kv10.1-expressing MDA-MB-435S cells. Moreover, using the ß-gal activatable NIRF probe DDAOG, we detected mAb62-ß-gal activity in vivo over the tumour area. In summary, we could show that the anti-Kv10.1 antibody is a promising tool for the development of novel concepts of targeted cancer therapy.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Transformación Celular Neoplásica , Canales de Potasio Éter-A-Go-Go/inmunología , Imagen Óptica/métodos , Animales , Carbocianinas/metabolismo , Línea Celular Tumoral , Canales de Potasio Éter-A-Go-Go/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones
5.
J Physiol ; 593(1): 181-96, 2015 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-25556795

RESUMEN

KEY POINTS: Voltage-gated KV 10.1 potassium channels are widely expressed in the mammalian brain but their function remains poorly understood. We report that KV 10.1 is enriched in the presynaptic terminals and does not take part in somatic action potentials. In parallel fibre synapses in the cerebellar cortex, we find that KV 10.1 regulates Ca(2+) influx and neurotransmitter release during repetitive high-frequency activity. Our results describe the physiological role of mammalian KV 10.1 for the first time and help understand the fine-tuning of synaptic transmission. The voltage-gated potassium channel KV 10.1 (Eag1) is widely expressed in the mammalian brain, but its physiological function is not yet understood. Previous studies revealed highest expression levels in hippocampus and cerebellum and suggested a synaptic localization of the channel. The distinct activation kinetics of KV 10.1 indicate a role during repetitive activity of the cell. Here, we confirm the synaptic localization of KV 10.1 both biochemically and functionally and that the channel is sufficiently fast at physiological temperature to take part in repolarization of the action potential (AP). We studied the role of the channel in cerebellar physiology using patch clamp and two-photon Ca(2+) imaging in KV 10.1-deficient and wild-type mice. The excitability and action potential waveform recorded at granule cell somata was unchanged, while Ca(2+) influx into axonal boutons was enhanced in mutants in response to stimulation with three APs, but not after a single AP. Furthermore, mutants exhibited a frequency-dependent increase in facilitation at the parallel fibre-Purkinje cell synapse at high firing rates. We propose that KV 10.1 acts as a modulator of local AP shape specifically during high-frequency burst firing when other potassium channels suffer cumulative inactivation.


Asunto(s)
Calcio/fisiología , Cerebelo/fisiología , Canales de Potasio Éter-A-Go-Go/fisiología , Canales de Potasio con Entrada de Voltaje/fisiología , Células de Purkinje/fisiología , Potenciales de Acción , Animales , Cerebelo/citología , Potenciales Postsinápticos Excitadores , Células HEK293 , Humanos , Ratones Noqueados , Ratas Sprague-Dawley , Sinapsis/fisiología
6.
Biochim Biophys Acta ; 1838(3): 921-31, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24269539

RESUMEN

KV10.1 potassium channels are implicated in a variety of cellular processes including cell proliferation and tumour progression. Their expression in over 70% of human tumours makes them an attractive diagnostic and therapeutic target. Although their physiological role in the central nervous system is not yet fully understood, advances in their precise cell localization will contribute to the understanding of their interactions and function. We have determined the plasma membrane (PM) distribution of the KV10.1 protein in an enriched mouse brain PM fraction and its association with cholesterol- and sphingolipid-rich domains. We show that the KV10.1 channel has two different populations in a 3:2 ratio, one associated to and another excluded from Detergent Resistant Membranes (DRMs). This distribution of KV10.1 in isolated PM is cholesterol- and cytoskeleton-dependent since alteration of those factors changes the relationship to 1:4. In transfected HEK-293 cells with a mutant unable to bind Ca(2+)/CaM to KV10.1 protein, Kv10.1 distribution in DRM/non-DRM is 1:4. Mean current density was doubled in the cholesterol-depleted cells, without any noticeable effects on other parameters. These results demonstrate that recruitment of the KV10.1 channel to the DRM fractions involves its functional regulation.


Asunto(s)
Membrana Celular/metabolismo , Canales de Potasio Éter-A-Go-Go/metabolismo , Microdominios de Membrana/metabolismo , Neuronas/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Animales , Western Blotting , Membrana Celular/química , Colesterol/metabolismo , Citoesqueleto/metabolismo , Detergentes/metabolismo , Electrofisiología , Canales de Potasio Éter-A-Go-Go/química , Canales de Potasio Éter-A-Go-Go/genética , Femenino , Células HEK293 , Humanos , Microdominios de Membrana/química , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Canales de Potasio con Entrada de Voltaje/química , Canales de Potasio con Entrada de Voltaje/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
BMC Cancer ; 15: 839, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26530050

RESUMEN

BACKGROUND: Kv10.1, a voltage-gated potassium channel only detected in the healthy brain, was found to be aberrantly expressed in extracerebral cancers. Investigations of Kv10.1 in brain metastasis and glioblastoma multiforme (GBM) are lacking. METHODS: We analyzed the expression of Kv10.1 by immunohistochemistry in these brain tumors (75 metastasis from different primary tumors, 71 GBM patients) and the influence of a therapy with tricyclic antidepressants (which are Kv10.1 blockers) on survival. We also investigated Kv10.1 expression in the corresponding primary carcinomas of metastases patients. RESULTS: We observed positive Kv10.1 expression in 85.3 % of the brain metastases and in 77.5 % of GBMs. Patients with brain metastases, showing low Kv10.1 expression, had a significantly longer overall survival compared to those patients with high Kv10.1 expression. Metastases patients displaying low Kv10.1 expression and also receiving tricyclic antidepressants showed a significantly longer median overall survival as compared to untreated patients. CONCLUSIONS: Our data show that Kv10.1 is not only highly expressed in malignant tumors outside CNS, but also in the most frequent cerebral cancer entities, metastasis and GBM, which remain incurable in spite of aggressive multimodal therapies. Our results extend the correlation between dismal prognosis and Kv10.1 expression to patients with brain metastases or GBMs and, moreover, they strongly suggest a role of tricyclic antidepressants for personalized therapy of brain malignancies.


Asunto(s)
Neoplasias Encefálicas/genética , Canales de Potasio Éter-A-Go-Go/biosíntesis , Glioblastoma/genética , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/secundario , Supervivencia sin Enfermedad , Canales de Potasio Éter-A-Go-Go/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , Humanos , Masculino , Persona de Mediana Edad , Pronóstico
8.
Life (Basel) ; 14(5)2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38792600

RESUMEN

Our research has developed a highly sensitive and simple assay to detect small amounts of animal and human biological material in less than 40 min. The handheld SaLux19 device developed at the Max Planck Institute of Experimental Medicine in Göttingen, Germany, was used to validate our concept. The proposed system uses isothermal amplification of DNA in a rapid assay format. Our results show that the assay can detect Sus scrofa nucleic acids with very high sensitivity and specificity. This detection system has potential for forensic scenarios.

9.
Glia ; 61(7): 1084-100, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23595698

RESUMEN

Neuron-glia interactions play a key role in maintaining and regulating the central nervous system. Glial cells are implicated in the function of dopamine neurons and regulate their survival and resistance to injury. Parkinson's disease is characterized by the loss of dopamine neurons in the substantia nigra pars compacta, decreased striatal dopamine levels and consequent onset of extrapyramidal motor dysfunction. Parkinson's disease is a common chronic, neurodegenerative disorder with no effective protective treatment. In the 6-OHDA mouse model of Parkinson's disease, doxycycline administered at a dose that both induces/represses conditional transgene expression in the tetracycline system, mitigates the loss of dopaminergic neurons in the substantia nigra compacta and nerve terminals in the striatum. This protective effect was associated with: (1) a reduction of microglia in normal mice as a result of doxycycline administration per se; (2) a decrease in the astrocyte and microglia response to the neurotoxin 6-OHDA in the globus pallidus and substantia nigra compacta, and (3) the astrocyte reaction in the striatum. Our results suggest that doxycycline blocks 6-OHDA neurotoxicity in vivo by inhibiting microglial and astrocyte expression. This action of doxycycline in nigrostriatal dopaminergic neuron protection is consistent with a role of glial cells in Parkinson's disease neurodegeneration. The neuroprotective effect of doxycycline may be useful in preventing or slowing the progression of Parkinson's disease and other neurodegenerative diseases linked to glia function.


Asunto(s)
Doxiciclina/uso terapéutico , Neuroglía/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/patología , Adrenérgicos/toxicidad , Análisis de Varianza , Animales , Encéfalo/metabolismo , Encéfalo/patología , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Antígeno de Macrófago-1/metabolismo , Masculino , Metaloproteinasa 3 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Oxidopamina/toxicidad , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/fisiopatología , Tirosina 3-Monooxigenasa/metabolismo
10.
EMBO Rep ; 11(6): 431-7, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20448665

RESUMEN

The prostate is a highly specialized mammalian organ that produces and releases large amounts of citrate. However, the citrate release mechanism is not known. Here, we present the results of molecular cloning of a citrate transporter from human normal prostate epithelial PNT2-C2 cells shown previously to express such a mechanism. By using rapid amplification of cDNA ends PCR, we determined that the prostatic carrier is an isoform of the mitochondrial transporter SLC25A1 with a different first exon. We confirmed the functionality of the clone by expressing it in human embryonic kidney cells and performing radiotracer experiments and whole-cell patch-clamp recordings. By using short interfering RNAs targeting different parts of the sequence, we confirmed that the cloned protein is the main prostatic transporter responsible for citrate release. We also produced a specific antibody and localized the cloned transporter protein to the plasma membrane of the cells. By using the same antibody, we have shown that the cloned transporter is expressed in non-malignant human tissues.


Asunto(s)
Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Células Epiteliales/metabolismo , Evolución Molecular , Próstata/citología , Secuencia de Aminoácidos , Proteínas de Transporte de Anión/química , Proteínas de Transporte de Anión/metabolismo , Transporte Biológico , Proteínas Portadoras/química , Línea Celular , Ácido Cítrico/metabolismo , Células Epiteliales/citología , Silenciador del Gen , Humanos , Inmunohistoquímica , Iones/metabolismo , Masculino , Proteínas Mitocondriales/química , Proteínas Mitocondriales/metabolismo , Datos de Secuencia Molecular , Transportadores de Anión Orgánico , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo
11.
Mol Biol Rep ; 39(3): 2187-94, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21643953

RESUMEN

Transfection has become an everyday technique widely used for functional studies in living cells. The choice of the particular transfection method is usually determined by its efficiency and toxicity, and possible functional consequences specific to the method used are normally overlooked. We describe here that nucleofection, a method increasingly used because of its convenience and high efficiency, increases the metabolic rate of some cancer cells, which can be misleading when used as a measure of proliferation. Moreover, nucleofection can alter the subcellular expression pattern of the transfected protein. These undesired effects are independent of the transfected nucleic acid, but depend on the particular cell line used. Therefore, the interpretation of functional data using this technology requires further controls and caution.


Asunto(s)
Núcleo Celular/metabolismo , Electroporación/métodos , Metabolismo Energético/fisiología , Transfección/métodos , Análisis de Varianza , Animales , Línea Celular , Proliferación Celular , Citometría de Flujo , Humanos , Ratones , Microscopía Fluorescente , Oxazinas/metabolismo , Oxidación-Reducción , Reacción en Cadena en Tiempo Real de la Polimerasa , Sales de Tetrazolio/metabolismo , Tiazoles/metabolismo , Xantenos/metabolismo
12.
Proteomics ; 11(2): 261-9, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21204253

RESUMEN

The molecular basis of bipolar disorder (BD) is still unknown as is the mechanism through which lithium, the therapy of choice, exerts its effects in treatment of BD. So far, no biomarkers exist to facilitate diagnosis of BD or treatment evaluation. To investigate whether BD and its treatment with lithium leaves a characteristic signature in the serum proteome, we used SELDI-TOF MS to analyze individual serum samples from BD patients treated with lithium (BD-plus-Li, n=15) or other drugs (BD-minus-Li, n=10) and from healthy controls (n=15). Interestingly, features of 28 kDa (one peak) and 14 kDa (three peaks) showed a decreased level in the BD-minus-Li group and a level restored to that of the control group in the BD-plus-Li group. To reveal the identity of these features, we subjected pooled serum samples from both BD groups to the 2-D DIGE technology and identified 28 kDa apolipoprotein A-I (apo A-I) and three 14 kDa fragments thereof as upregulated in the BD-plus-Li group. Immunoturbidimetry, a routine clinical assay, verified the characteristic apo A-I signature in individual serum samples. In conclusion, we propose apo A-I as a candidate marker that can visualize response to lithium treatment at the serum protein level.


Asunto(s)
Antipsicóticos/uso terapéutico , Apolipoproteína A-I/sangre , Trastorno Bipolar/diagnóstico , Trastorno Bipolar/tratamiento farmacológico , Compuestos de Litio/uso terapéutico , Adulto , Secuencia de Aminoácidos , Biomarcadores/sangre , Femenino , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Proteoma/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
13.
Mol Cancer ; 10: 109, 2011 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-21899742

RESUMEN

BACKGROUND: The search for strategies to target ion channels for therapeutic applications has become of increasing interest. Especially, the potassium channel K(V)10.1 (Ether-á-go-go) is attractive as target since this surface protein is virtually not detected in normal tissue outside the central nervous system, but is expressed in approximately 70% of tumors from different origins. METHODS: We designed a single-chain antibody against an extracellular region of K(V)10.1 (scFv62) and fused it to the human soluble TRAIL. The K(V)10.1-specific scFv62 antibody -TRAIL fusion protein was expressed in CHO-K1 cells, purified by chromatography and tested for biological activity. RESULTS: Prostate cancer cells, either positive or negative for K(V)10.1 were treated with the purified construct. After sensitization with cytotoxic drugs, scFv62-TRAIL induced apoptosis only in K(V)10.1-positive cancer cells, but not in non-tumor cells, nor in tumor cells lacking K(V)10.1 expression. In co-cultures with K(V)10.1-positive cancer cells the fusion protein also induced apoptosis in bystander K(V)10.1-negative cancer cells, while normal prostate epithelial cells were not affected when present as bystander. CONCLUSIONS: K(V)10.1 represents a novel therapeutic target for cancer. We could design a strategy that selectively kills tumor cells based on a K(V)10.1-specific antibody.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Biomarcadores de Tumor/metabolismo , Canales de Potasio Éter-A-Go-Go/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Anticuerpos de Cadena Única/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Antineoplásicos/aislamiento & purificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Biomarcadores de Tumor/genética , Efecto Espectador , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cicloheximida/farmacología , Canales de Potasio Éter-A-Go-Go/genética , Expresión Génica/efectos de los fármacos , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/aislamiento & purificación , Anticuerpos de Cadena Única/biosíntesis , Anticuerpos de Cadena Única/aislamiento & purificación , Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Ligando Inductor de Apoptosis Relacionado con TNF/aislamiento & purificación
14.
Mol Cancer ; 9: 18, 2010 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-20105281

RESUMEN

BACKGROUND: The voltage-gated potassium channel hEag1 (KV10.1) has been related to cancer biology. The physiological expression of the human channel is restricted to the brain but it is frequently and abundantly expressed in many solid tumors, thereby making it a promising target for a specific diagnosis and therapy. Because chronic lymphatic leukemia has been described not to express hEag1, it has been assumed that the channel is not expressed in hematopoietic neoplasms in general. RESULTS: Here we show that this assumption is not correct, because the channel is up-regulated in myelodysplastic syndromes, chronic myeloid leukemia and almost half of the tested acute myeloid leukemias in a subtype-dependent fashion. Most interestingly, channel expression strongly correlated with increasing age, higher relapse rates and a significantly shorter overall survival. Multivariate Cox regression analysis revealed hEag1 expression levels in AML as an independent predictive factor for reduced disease-free and overall survival; such an association had not been reported before. As a functional correlate, specific hEag1 blockade inhibited the proliferation and migration of several AML cell lines and primary cultured AML cells in vitro. CONCLUSION: Our observations implicate hEag1 as novel target for diagnostic, prognostic and/or therapeutic approaches in AML.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/metabolismo , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/fisiopatología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento/patología , Línea Celular Tumoral , Niño , Preescolar , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/genética , Femenino , Regulación Leucémica de la Expresión Génica , Humanos , Lactante , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Masculino , Persona de Mediana Edad , Pronóstico , Análisis de Supervivencia , Resultado del Tratamiento , Adulto Joven
15.
BMC Psychiatry ; 10: 91, 2010 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-21067598

RESUMEN

BACKGROUND: Schizophrenia is the collective term for an exclusively clinically diagnosed, heterogeneous group of mental disorders with still obscure biological roots. Based on the assumption that valuable information about relevant genetic and environmental disease mechanisms can be obtained by association studies on patient cohorts of ≥ 1000 patients, if performed on detailed clinical datasets and quantifiable biological readouts, we generated a new schizophrenia data base, the GRAS (Göttingen Research Association for Schizophrenia) data collection. GRAS is the necessary ground to study genetic causes of the schizophrenic phenotype in a 'phenotype-based genetic association study' (PGAS). This approach is different from and complementary to the genome-wide association studies (GWAS) on schizophrenia. METHODS: For this purpose, 1085 patients were recruited between 2005 and 2010 by an invariable team of traveling investigators in a cross-sectional field study that comprised 23 German psychiatric hospitals. Additionally, chart records and discharge letters of all patients were collected. RESULTS: The corresponding dataset extracted and presented in form of an overview here, comprises biographic information, disease history, medication including side effects, and results of comprehensive cross-sectional psychopathological, neuropsychological, and neurological examinations. With >3000 data points per schizophrenic subject, this data base of living patients, who are also accessible for follow-up studies, provides a wide-ranging and standardized phenotype characterization of as yet unprecedented detail. CONCLUSIONS: The GRAS data base will serve as prerequisite for PGAS, a novel approach to better understanding 'the schizophrenias' through exploring the contribution of genetic variation to the schizophrenic phenotypes.


Asunto(s)
Recolección de Datos/métodos , Fenotipo , Esquizofrenia/genética , Adolescente , Adulto , Anciano , Antipsicóticos/efectos adversos , Antipsicóticos/uso terapéutico , Enfermedades de los Ganglios Basales/inducido químicamente , Enfermedades de los Ganglios Basales/diagnóstico , Trastornos del Conocimiento/diagnóstico , Trastornos del Conocimiento/psicología , Estudios Transversales , Bases de Datos Genéticas/estadística & datos numéricos , Femenino , Estudios de Asociación Genética , Humanos , Masculino , Persona de Mediana Edad , Pruebas Neuropsicológicas , Esquizofrenia/diagnóstico , Esquizofrenia/tratamiento farmacológico , Psicología del Esquizofrénico
16.
Eur Biophys J ; 38(3): 279-84, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18379771

RESUMEN

The ether-à-go-go potassium channels hEag1 and hEag2 are highly homologous. Even though both possess identical voltage-sensing domain S4, the channels act differently in response to voltage. Therefore we asked whether transmembrane domains other than the voltage sensor could contribute to the voltage-dependent behaviour of these potassium channels. For this chimaeras were created, in which each single transmembrane domain of hEag1 was replaced by the corresponding segment of hEag2. The voltage-dependent properties of the chimaeras were analysed after expression in Xenopus laevis oocytes using the two-electrode voltage-clamp method. By this we found, that only the mutations in transmembrane domains S5 and S6 are able to change the voltage sensitivity of hEag1 by shifting the half-activation potential (V(50)) to values intermediate between the two wild types. Moreover, the presence of Mg2+ has strong effects on the voltage sensitivity of hEag2 shifting V(50) by more than 50 mV to more positive values. Interestingly, despite the identical binding site Mg2+ showed only little effects on hEag1 or the chimaeras. Altogether, our data suggest that not only transmembrane spanning regions, but also non-membrane spanning regions are responsible for differences in the behaviour of the hEag1 and hEag2 potassium channels.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/fisiología , Animales , Membrana Celular/fisiología , Canales de Potasio Éter-A-Go-Go/genética , Humanos , Activación del Canal Iónico , Magnesio/metabolismo , Mutagénesis Sitio-Dirigida , Oocitos/fisiología , Técnicas de Placa-Clamp , Estructura Terciaria de Proteína , Xenopus laevis
17.
Cancer Res ; 67(15): 7343-9, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17671204

RESUMEN

The potassium channel ether à go-go has been directly linked to cellular proliferation and transformation, although its physiologic role(s) are as of yet unknown. The specific blockade of human Eag1 (hEag1) may not only allow the dissection of the role of the channel in distinct physiologic processes, but because of the implication of hEag1 in tumor biology, it may also offer an opportunity for the treatment of cancer. However, members of the potassium channel superfamily are structurally very similar to one another, and it has been notoriously difficult to obtain specific blockers for any given channel. Here, we describe and validate the first rational design of a monoclonal antibody that selectively inhibits a potassium current in intact cells. Specifically blocking hEag1 function using this antibody inhibits tumor cell growth both in vitro and in vivo. Our data provide a proof of concept that enables the generation of functional antagonistic monoclonal antibodies against ion channels with therapeutic potential. The particular antibody described here, as well as the technique developed to make additional functional antibodies to Eag1, makes it possible to evaluate the potential of the channel as a target for cancer therapy.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Canales de Potasio Éter-A-Go-Go/inmunología , Neoplasias Mamarias Experimentales/terapia , Neoplasias Pancreáticas/terapia , Bloqueadores de los Canales de Potasio/uso terapéutico , Animales , Proliferación Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Electrofisiología , Ensayo de Inmunoadsorción Enzimática , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Riñón/citología , Riñón/metabolismo , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones SCID , Mutagénesis Sitio-Dirigida , Neuroblastoma/inmunología , Neuroblastoma/metabolismo , Neuroblastoma/patología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/metabolismo , Trasplante Heterólogo
18.
Nat Neurosci ; 6(8): 846-53, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12845327

RESUMEN

Ca2+ microdomains that form during the opening of voltage-gated Ca2+ channels have been implicated in regulating the kinetics of hormone and transmitter release. Direct assessment of the interaction between a single Ca2+ microdomain and a single secretory vesicle has been impossible because of technical limitations. Using evanescent field imaging of near-membrane micromolar Ca2+ concentration ([Ca2+]) and fluorescently labeled vesicles, we have observed exocytosis of individual chromaffin dense-core vesicles that was triggered by Ca2+ microdomains. Ca2+ microdomains selectively triggered the release of vesicles that were docked within 300 nm. Not all vesicles exposed to a Ca2+ microdomain were released, indicating that some vesicles are docked but are not ready for release. In addition to its established role as a trigger for release, elevated near-membrane [Ca2+] reduced the distance between docked vesicles and Ca2+ entry sites. Our results suggest a new mechanism for stimulation-dependent facilitation of exocytosis, whereby vesicles are moved closer to Ca2+ entry sites, thereby increasing a Ca2+ microdomain's efficacy to trigger vesicle fusion.


Asunto(s)
Canales de Calcio/fisiología , Calcio/fisiología , Exocitosis/fisiología , Vesículas Secretoras/metabolismo , Potenciales de Acción , Calcio/metabolismo , Canales de Calcio/química , Células Cultivadas , Células Cromafines/metabolismo , Concentración Osmolar , Estructura Terciaria de Proteína/fisiología
19.
Toxicon ; 49(4): 452-72, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17215013

RESUMEN

Receptor site-3 on voltage-gated sodium channels is targeted by a variety of structurally distinct toxins from scorpions, sea anemones, and spiders whose typical action is the inhibition of sodium current inactivation. This site interacts allosterically with other topologically distinct receptors that bind alkaloids, lipophilic polyether toxins, pyrethroids, and site-4 scorpion toxins. These features suggest that design of insecticides with specificity for site-3 might be rewarding due to the positive cooperativity with other toxins or insecticidal agents. Yet, despite the central role of scorpion alpha-toxins in envenomation and their vast use in the study of channel functions, molecular details on site-3 are scarce. Scorpion alpha-toxins vary greatly in preference for sodium channels of insects and mammals, and some of them are highly active on insects. This implies that despite its commonality, receptor site-3 varies on insect vs. mammalian channels, and that elucidation of these differences could potentially be exploited for manipulation of toxin preference. This review provides current perspectives on (i) the classification of scorpion alpha-toxins, (ii) their mode of interaction with sodium channels and pharmacological divergence, (iii) molecular details on their bioactive surfaces and differences associated with preference for channel subtypes, as well as (iv) a summary of the present knowledge about elements involved in constituting receptor site-3. These details, combined with the variations in allosteric interactions between site-3 and the other receptor sites on insect and mammalian sodium channels, may be useful in new strategies of insect control and future design of anti-insect selective ligands.


Asunto(s)
Neurotoxinas/toxicidad , Control Biológico de Vectores , Venenos de Escorpión/toxicidad , Escorpiones/fisiología , Canales de Sodio/efectos de los fármacos , Fosfolipasas de Tipo C/toxicidad , Secuencia de Aminoácidos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Saltamontes , Datos de Secuencia Molecular , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neurotoxinas/química , Conformación Proteica , Ratas , Venenos de Escorpión/química , Canales de Sodio/química , Canales de Sodio/metabolismo , Fosfolipasas de Tipo C/química
20.
Toxicon ; 49(4): 473-89, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17197009

RESUMEN

Voltage-gated sodium channels are a major target for toxins and insecticides due to their central role in excitability, but due to the conservation of these channels in Animalia most insecticides do not distinguish between those of insects and mammals, thereby imposing risks to humans and livestock. Evidently, as long as modern agriculture depends heavily on the use of insecticides there is a great need for new substances capable of differentiating between sodium channel subtypes. Such substances exist in venomous animals, but ways for their exploitation have not yet been developed due to problems associated with manufacturing, degradation, and delivery to the target channels. Engineering of plants for expression of anti-insect toxins or use of natural vectors that express toxins near their target site (e.g. baculoviruses) are still problematic and raise public concern. In this problematic reality a rational approach might be to learn from nature how to design highly selective anti-insect compounds preferably in the form of peptidomimetics. This is a complex task that requires the elucidation of the face of interaction between insect-selective toxins and their sodium channel receptor sites. This review delineates current progress in: (i) elucidation of the bioactive surfaces of scorpion beta-toxins, especially the excitatory and depressant groups, which show high preference for insects and bind insect sodium channels with high affinity; (ii) studies of the mode of interaction of scorpion beta-toxins with receptor site-4 on voltage-gated sodium channels; and (iii) clarification of channel elements that constitute receptor site-4. This information may be useful in future attempts to mimic the bioactive surface of the toxins for the design of anti-insect selective peptidomimetics.


Asunto(s)
Proteínas de Insectos/toxicidad , Neurotoxinas/toxicidad , Control Biológico de Vectores , Venenos de Escorpión/toxicidad , Escorpiones/fisiología , Canales de Sodio/efectos de los fármacos , Agricultura/métodos , Secuencia de Aminoácidos , Animales , Proteínas de Insectos/química , Datos de Secuencia Molecular , Neurotoxinas/química , Conformación Proteica , Venenos de Escorpión/química , Canales de Sodio/química , Canales de Sodio/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA